1
|
Jia M, Li Y, Wang C, Gao X, Guan Y, Ai H. Fluorescence Detection and Inhibition Mechanisms of DNTPH on Aβ42 Oligomers Characterized as Products in the Four Stages of Aggregation. ACS Chem Neurosci 2024. [PMID: 39494683 DOI: 10.1021/acschemneuro.4c00509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024] Open
Abstract
Aβ42 aggregation was implicated in the pathogenesis of Alzheimer's disease (AD) without effective treatment available currently. Future efforts in clinical trials should instead focus on applying those antiamyloid treatment strategies to the preclinical stage and "the earlier, the better". How to identify and inhibit Aβ42 oligomers in the different stages of aggregation is therefore becoming the key to controlling primary aggregation and consequent AD development. Aggregation-induced emission probe DNTPH was demonstrated recently, enabling detection of amyloid at wavelengths up to 710 nm and exhibiting strong inhibitory effects on Aβ fibrosis at low dose. However, the detection and inhibition mechanisms of Aβ oligomers at various early stages of aggregation remain unknown. To this end, we built four different morphologies of Aβ42 pentamers characterized by products in monomeric aggregate (PM), primary nucleation (PP), secondary nucleation (PS), and fibril stages (PF) to explore the distinguishable ability and inhibition mechanisms of DNTPH with different concentrations upon binding. The results showcased that DNTPH does detect the four different Aβ42 oligomers with conspicuous fluorescence (λPM = 657 nm, λPP = 639 nm, λPS = 630 nm, and λPF = 648 nm) but fails to distinguish them, indicating that additional improvements are required further for the probe to achieve it. The inhibition mechanisms of DNTPH on the four Aβ42 aggregation are however of amazing differences. For PM and PP, aggregation was inhibited by altering the secondary structural composition, i.e., by decreasing the β-sheet and toxic turn (residues 22-23) probabilities, respectively. For PS, inhibition was achieved by segregating and keeping the two disordered monomeric species (PSM) away from the ordered secondary seed species (PSF) and consequently blocking further growth of the PSF seed. The inhibition mechanism for PS is first probed and proposed so far, as far as we know, and the corresponding aggregation stage of PS is the most important one among the four stages. The inhibition of PF was triggered by distorting the fibril chains, disrupting the ordered fibril surface for the contact of monomers. In addition, the optimal inhibitory concentrations of DNTPH for PM, PP, and PF were determined to be 1:3, while for PS, it was 1:5. This outcome offers a novel perspective for designing drugs targeting Aβ42 oligomers at different aggregation stages.
Collapse
Affiliation(s)
- Mengke Jia
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
- Zibo City Engineering Research Center for New Pollution Monitoring and Governance, Shandong Vocational College of Light Industry, Zibo, Shandong 255300, China
| | - Ye Li
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Chuanbo Wang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Xvzhi Gao
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Yvning Guan
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Hongqi Ai
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| |
Collapse
|
2
|
Zarrilli B, Bonanni R, Belfiore M, Severino M, Cariati I, Fioravanti R, Cappella G, Sennato S, Frank C, Giordani C, Tancredi V, Bombelli C, Diociaiuti M, D'Arcangelo G. Molecular mechanisms at the basis of the protective effect exerted by EPPS on neurodegeneration induced by prefibrillar amyloid oligomers. Sci Rep 2024; 14:26533. [PMID: 39489758 PMCID: PMC11532462 DOI: 10.1038/s41598-024-77859-9] [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: 05/31/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024] Open
Abstract
It has been shown recently, without an explanation of the possible molecular mechanisms involved, that 4-(2-hydroxyethyl)-1-piperazinepropanesulphonic (EPPS) acid effectively protects from the neurotoxicity induced by oligomers and plaques formed by the protein amyloid-β protein. Here we report the same protective effect, obtained in vitro (HT22-diff cell line) and ex vivo (hippocampal slices) models, against amyloid neurotoxicity induced by oligomers of salmon Calcitonin (sCT), which has been shown to be a good model for the study of neurodegenerative diseases. Based on biophysical studies focusing on the protein aggregation kinetic and the interaction of the aggregates with model membranes, we propose a possible molecular mechanism underlying the protective effects. Taken together, our results indicate that EPPS is able to counteract the direct association (primary aggregation) of harmless low-molecular weight aggregates (dimers and trimers) or their aggregation catalysed by surfaces present in the solution (secondary aggregation). Thus, EPPS stabilizes harmless aggregates and hinders the formation of toxic and metastable prefibrillar oligomers. Overall, our data demonstrate that EPPS is an excellent drug candidate for the treatment of neurodegeneration due to misfolded proteins, such as Alzheimer's or Parkinson's disease.
Collapse
Affiliation(s)
- Beatrice Zarrilli
- Department of Systems Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
- Laboratory of Experimental Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Roberto Bonanni
- Department of Biomedicine and Prevention, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
| | - Marcello Belfiore
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Mariagrazia Severino
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
| | - Raoul Fioravanti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Giacomo Cappella
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Sennato
- CNR - Institute of Complex Systems (ISC) - Sede "Sapienza", c/o Physics Department, "Sapienza" University of Rome, 00185, Rome, Italy
| | - Claudio Frank
- UniCamillus - Saint Camillus International University of Health Sciences, Via di Sant'Alessandro 8, Rome, 00131, Italy
| | - Cristiano Giordani
- Instituto de Física, Universidad de Antioquia, Calle 70 No. 52-21, 050010, Medellín, Colombia
- Grupo Productos Naturales Marinos, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Calle 70 No. 52-21, 050010, Medellín, Colombia
| | - Virginia Tancredi
- Department of Systems Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
- Centre of Space Bio-Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
| | - Cecilia Bombelli
- CNR - Institute for Biological Systems, Secondary Office of Rome - Reaction Mechanisms c/o Chemistry Department, "Sapienza" University of Rome, Rome, Italy
| | - Marco Diociaiuti
- CNR - Institute for Biological Systems, Secondary Office of Rome - Reaction Mechanisms c/o Chemistry Department, "Sapienza" University of Rome, Rome, Italy.
| | - Giovanna D'Arcangelo
- Department of Systems Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
- Centre of Space Bio-Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
| |
Collapse
|
3
|
Wu J, Wu J, Chen T, Cai J, Ren R. Protein aggregation and its affecting mechanisms in neurodegenerative diseases. Neurochem Int 2024; 180:105880. [PMID: 39396709 DOI: 10.1016/j.neuint.2024.105880] [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: 07/22/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
Protein aggregation serves as a critical pathological marker in a spectrum of neurodegenerative diseases (NDs), including the formation of amyloid β (Aβ) and Tau neurofibrillary tangles in Alzheimer's disease, as well as α-Synuclein (α-Syn) aggregates in Parkinson's disease, Parkinson's disease-related dementia (PDD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). A significant proportion of patients with amyotrophic lateral sclerosis (ALS) exhibit TDP-43 aggregates. Moreover, a confluence of brain protein pathologies, such as Aβ, Tau, α-Syn, and TDP-43, has been identified in individual NDs cases, highlighting the intricate interplay among these proteins that is garnering heightened scrutiny. Importantly, protein aggregation is modulated by an array of factors, with burgeoning evidence suggesting that it frequently results from perturbations in protein homeostasis, influenced by the cellular membrane milieu, metal ion concentrations, post-translational modifications, and genetic mutations. This review delves into the pathological underpinnings of protein aggregation across various NDs and elucidates the intercommunication among disparate proteins within the same disease context. Additionally, we examine the pathogenic mechanisms by which diverse factors impinge upon protein aggregation, offering fresh perspectives for the future therapeutic intervention of NDs.
Collapse
Affiliation(s)
- Junyun Wu
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jianan Wu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Tao Chen
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jing Cai
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| | - Reng Ren
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
4
|
Porel P, Bala K, Aran KR. Exploring the role of HIF-1α on pathogenesis in Alzheimer's disease and potential therapeutic approaches. Inflammopharmacology 2024:10.1007/s10787-024-01585-x. [PMID: 39465478 DOI: 10.1007/s10787-024-01585-x] [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: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
Hypoxia-inducible factor 1α (HIF-1α) is a crucial transcription factor that regulates cellular responses to low oxygen levels (hypoxia). In Alzheimer's disease (AD), emerging evidence suggests a significant involvement of HIF-1α in disease pathogenesis. AD is characterized by the accumulation of amyloid-beta (Aβ) plaques and neurofibrillary tangles (NFTs), leading to neuronal dysfunction and cognitive decline. HIF-1α is implicated in AD through its multifaceted roles in various cellular processes. Firstly, in response to hypoxia, HIF-1α promotes the expression of genes involved in angiogenesis, which is crucial for maintaining cerebral blood flow and oxygen delivery to the brain. However, in the context of AD, dysregulated HIF-1α activation may exacerbate cerebral hypoperfusion, contributing to neuronal damage. Moreover, HIF-1α is implicated in the regulation of Aβ metabolism. It can influence the production and clearance of Aβ peptides, potentially modulating their accumulation and toxicity in the brain. Additionally, HIF-1α activation has been linked to neuroinflammation, a key feature of AD pathology. It can promote the expression of pro-inflammatory cytokines and exacerbate neuronal damage. Furthermore, HIF-1α may play a role in synaptic plasticity and neuronal survival, which are impaired in AD. Dysregulated HIF-1α signaling could disrupt these processes, contributing to cognitive decline and neurodegeneration. Overall, the involvement of HIF-1α in various aspects of AD pathophysiology highlights its potential as a therapeutic target. Modulating HIF-1α activity could offer novel strategies for mitigating neurodegeneration and preserving cognitive function in AD patients. However, further research is needed to elucidate the precise mechanisms underlying HIF-1α dysregulation in AD and to develop targeted interventions.
Collapse
Affiliation(s)
- Pratyush Porel
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Kanchan Bala
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Khadga Raj Aran
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
5
|
Dhiman N, Deshwal S, Rishi V, Singhal N, Sandhir R. Zebrafish as a model organism to study sporadic Alzheimer's disease: Behavioural, biochemical and histological validation. Exp Neurol 2024; 383:115034. [PMID: 39490623 DOI: 10.1016/j.expneurol.2024.115034] [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: 06/28/2024] [Revised: 10/10/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) is a global burden to the healthcare system with no viable treatment options till date. Rodents and primates have been extensively used as models for understanding AD pathogenesis and identifying therapeutic targets. However, the focus is now shifting towards developing alternate models. Zebrafish is emerging as a preferred model for neurodegenerative conditions because of its simple nervous system, highly conserved genome and short duration required to model disease condition. The present study is aimed to develop streptozotocin (STZ)-induced model of sporadic AD (sAD) in zebrafish. STZ was administered to adult zebrafish (4-6 mo) at different doses (1 to 50 mg/kg body weight, intracerebroventricularly). Kaplan-Meier survival analysis revealed time and dose dependent mortality in the zebrafish administered with STZ. Based on survival analysis, 1 to 10 mg/kg body weight of STZ was selected for behavioural, molecular and histological studies. STZ administered fish had anxiety and stress-like behaviour in novel tank and light/dark preference tests. STZ-induced cognitive and memory deficits assessed using novel object recognition and spatial alternation tests. Further, expression of markers of amyloidogenic pathway (appa and bace1) were increased in terms of mRNA and protein levels in a time and dose dependent manner following STZ administration. However, expression of non-amyloidogenic pathway mediator (adam10) was reduced at both mRNA and protein level. Histological assessment using hematoxylin and eosin, and Nissl stain revealed loss of neurons in STZ administered fish. The ratio of phosphor-tauser396/total-tau was increased in STZ administered fish. Based on these findings, 5 mg/kg body weight of STZ was found to be most appropriate dose to exhibit sAD phenotype. Mass spectrometric analysis confirmed the presence of amyloid beta oligomers in brains of STZ administered fish. Transmission electron microscopy also showed the presence of higher order insoluble amyloid fibrils with twists. Immunohistochemical analysis revealed amyloid beta deposits in brain of STZ administered fish. Golgi-cox staining indicated decreased number of dendrites, whereas microglia had increased density, span ratio, soma area and lacunarity. The results of the present study demonstrate presence of AD hallmarks and phenotype in zebrafish 7 days post STZ administration (5 mg/kg). The study validates the potential of STZ-induced sAD in zebrafish as a reliable model for studying pathophysiology and rapid screening of therapeutic molecules against sAD.
Collapse
Affiliation(s)
- Neha Dhiman
- Department of Biochemistry, Hargobind Khorana Block, Panjab University, Sector 25, Chandigarh 160014, India
| | - Sonam Deshwal
- Department of Biochemistry, Hargobind Khorana Block, Panjab University, Sector 25, Chandigarh 160014, India
| | - Vikas Rishi
- National Agri-Food Biotechnology Institute, Sector 81, Mohali, Punjab 140306, India
| | - Nitin Singhal
- National Agri-Food Biotechnology Institute, Sector 81, Mohali, Punjab 140306, India
| | - Rajat Sandhir
- Department of Biochemistry, Hargobind Khorana Block, Panjab University, Sector 25, Chandigarh 160014, India.
| |
Collapse
|
6
|
Hatakawa Y, Takeuchi Y, Lee SH, Oe T. Tyrosine modifications of insulin-degrading enzyme enable favorable control of substrate specificity for both Alzheimer's disease and type-2 diabetes mellitus. Bioorg Chem 2024; 153:107916. [PMID: 39481143 DOI: 10.1016/j.bioorg.2024.107916] [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: 06/21/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024]
Abstract
Insulin-degrading enzyme (IDE) cleaves amyloid beta (Aβ), insulin, and other bioactive peptides. Because Aβ and insulin are closely related to Alzheimer's disease (AD) and type-2 diabetes mellitus (T2DM), respectively, IDE is a candidate drug target for treating both AD and T2DM. However, the activity of IDE has opposing effects, including decreasing AD risk by degrading Aβ and increasing T2DM risk by degrading insulin. The opposed substrate specificity is associated with the exo- and active sites containing Tyr314 and Tyr831 residues, the plausible modification targets for controlling substrate specificity. In this study, we used a tyrosine-specific modification regent, Cookson reagent (4-phenyl-1,2,4-triazoline-3,5-dione, PTAD), for IDE and examined the degradation activities on Aβ40 and insulin. Fifteen tyrosine residues, including Tyr314 and Tyr831, were modified by PTAD. After incubation with PTAD-modified IDE for 3 days, insulin remained intact, whereas Aβ40 was completely degraded. This favorable change of substrate specificity was also observed in the mixture of Aβ40 and insulin, suggesting that tyrosine modification of IDE might be a therapeutic strategy for AD and T2DM.
Collapse
Affiliation(s)
- Yusuke Hatakawa
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan
| | - Yuki Takeuchi
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan
| | - Seon Hwa Lee
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan
| | - Tomoyuki Oe
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan.
| |
Collapse
|
7
|
Mackness BC, Morgan BR, Deveau LM, Kathuria SV, Zitzewitz JA, Massi F. A Hydrophobic Core Stabilizes the Residual Structure in the RRM2 Intermediate State of the ALS-linked Protein TDP-43. J Mol Biol 2024; 436:168823. [PMID: 39426615 DOI: 10.1016/j.jmb.2024.168823] [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: 07/24/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Folding intermediates mediate both protein folding and the misfolding and aggregation observed in human diseases, including amyotrophic lateral sclerosis (ALS), and are prime targets for therapeutic interventions. In this study, we identified the core nucleus of structure for a folding intermediate in the second RNA recognition motif (RRM2) of the ALS-linked RNA-binding protein, TDP-43 (TAR DNA-binding protein-43), using a combination of experimental and computational approaches. Urea equilibrium unfolding studies revealed that the RRM2 intermediate state consists of collapsed residual secondary structure localized to the N-terminal half of RRM2, while the C-terminus is largely disordered. Steered molecular dynamics simulations and mutagenesis studies yielded key stabilizing hydrophobic contacts that, when mutated to alanine, severely disrupt the overall fold of RRM2. In combination, these findings suggest a role for this RRM intermediate in normal TDP-43 function as well as serving as a template for misfolding and aggregation through the low stability and non-native secondary structure.
Collapse
Affiliation(s)
- Brian C Mackness
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Brittany R Morgan
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Laura M Deveau
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sagar V Kathuria
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jill A Zitzewitz
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.
| | - Francesca Massi
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
8
|
Menendez-Gonzalez M. Intrathecal Immunoselective Nanopheresis for Alzheimer's Disease: What and How? Why and When? Int J Mol Sci 2024; 25:10632. [PMID: 39408961 PMCID: PMC11476806 DOI: 10.3390/ijms251910632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
Nanotechnology is transforming therapeutics for brain disorders, especially in developing drug delivery systems. Intrathecal immunoselective nanopheresis with soluble monoclonal antibodies represents an innovative approach in the realm of drug delivery systems for Central Nervous System conditions, especially for targeting soluble beta-amyloid in Alzheimer's disease. This review delves into the concept of intrathecal immunoselective nanopheresis. It provides an overall description of devices to perform this technique while discussing the nanotechnology behind its mechanism of action, its potential advantages, and clinical implications. By exploring current research and advancements, we aim to provide a comprehensive understanding of this novel method, addressing the critical questions of what it is, how it works, why it is needed, and when it should be applied. Special attention is given to patient selection and the optimal timing for therapy initiation in Alzheimer's, coinciding with the peak accumulation of amyloid oligomers in the early stages. Potential limitations and alternative targets beyond beta-amyloid and future perspectives for immunoselective nanopheresis are also described.
Collapse
Affiliation(s)
- Manuel Menendez-Gonzalez
- Departamento de Medicina, Facultad de Ciencias de la Salud, Universidad de Oviedo, ES-33006 Oviedo, Spain;
- Hospital Universitario Central de Asturias, Servicio de Neurología, ES-33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), ES-33011 Oviedo, Spain
| |
Collapse
|
9
|
Zahraee H, Mohammadi F, Parvaee E, Khoshbin Z, Arab SS. Reducing the assemblies of amyloid-beta multimers by sodium dodecyl sulfate surfactant at concentrations lower than critical micelle concentration: molecular dynamics simulation exploration. J Biomol Struct Dyn 2024; 42:8673-8687. [PMID: 37599504 DOI: 10.1080/07391102.2023.2247086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/05/2023] [Indexed: 08/22/2023]
Abstract
Amyloid-β peptide, the predominant proteinaceous component of senile plaques, is responsible for the incidence of Alzheimer's disease (AD), an age-associated neurodegenerative disorder. Specifically, the amyloid-β(1-42) (Aβ1-42) isoform, known for its high toxicity, is the predominant biomarker for the preliminary diagnosis of AD. The aggregation of the Aβ1-42 peptides can be affected by the components of the cellular medium through changing their structures and molecular interactions. In this study, we investigated the effect of sodium dodecyl sulfate (SDS) at much lower concentrations than the critical micelle concentration (CMC) on Aβ1-42 aggregation. For this purpose, we studied mono-, di-, tri- and tetramers of Aβ1-42 peptide in two different concentrations of SDS molecules (10 and 40 molecules) using a 300 ns molecular dynamics simulation for each system. The distance between the center of mass (COM) of Aβ1-42 peptides confirms that an increase in the number of SDS molecules decreases their aggregation probability due to greater interaction with SDS molecules. Besides, the less compactness parameter reveals the reduced aggregation probability of Aβ1-42 peptides. Based on the energetic FEL landscapes, SDS molecules with the concentration closer to the CMC are an effective inhibitory agent to prevent the formation of Aβ1-42 fibrils. Also, the aggregation direction of the peptide pairs can be predicted by determining the direction of the accumulation-deterrent forces.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Hamed Zahraee
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mohammadi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elahe Parvaee
- Department of Chemistry, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Zahra Khoshbin
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Shahriar Arab
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
10
|
Huf F, Gutierres JM, da Silva GN, Zago AM, Koenig LFC, Fernandes MC. Neuroprotection elicited by taurine in sporadic Alzheimer-like disease: benefits on memory and control of neuroinflammation in the hippocampus of rats. Mol Cell Biochem 2024; 479:2663-2678. [PMID: 37874493 DOI: 10.1007/s11010-023-04872-3] [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: 07/06/2023] [Accepted: 10/05/2023] [Indexed: 10/25/2023]
Abstract
This study aimed to analyze whether taurine has a nootropic effect on short-term and long-term memory in a model of sporadic dementia of the Alzheimer's type (SDAT). Moreover, we evaluated the immunoreactivity and insulin receptor (IR) distribution and markers for neurons and glial cells in the hippocampus of rats with SDAT and treated with taurine. For this, Male Wistar rats received STZ (ICV, 3 mg/kg, bilateral, 5ul per site, aCFS vehicle) and were treated with taurine (100 mg/kg orally, 1 time per day, saline vehicle) for 25 days. The animals were divided into 4 groups: vehicle (VE), taurine (TAU), ICV-STZ (STZ) and ICV-STZ plus taurine (STZ + TAU). At the end of taurine treatment, short- and long-term memory were assessed by performance on object recognition and Y-maze tasks. Insulin receptor (IR) was evaluated by immunoperoxidase while mature neurons (NeuN), astrocytes (GFAP, S100B, SOX9), and microglia (Iba-1) were evaluated by immunofluorescence. STZ induced worse spatial and recognition memory (INDEX) in YM and ORT tasks. Taurine protected against STZ-induced memory impairment. SDAT reduced the population of mature neurons as well as increased astrocytic and microglial reactivity, and taurine protected against these STZ-induced effects, mainly in the CA1 region of the hippocampus. Taurine increases IR expression in the hippocampus, and protects against the reduction in the density of this receptor in CA1 induced by STZ. In conclusion, these findings demonstrate that taurine is able to enhance memory, up-regulates IR in the hippocampus, protects the neuron population, and reduces the astrogliosis found in SDAT.
Collapse
Affiliation(s)
- Fernanda Huf
- Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514 - Building 3, Porto Alegre, CEP 90050-170, RS, Brazil
| | - Jessié Martins Gutierres
- Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514 - Building 3, Porto Alegre, CEP 90050-170, RS, Brazil.
| | - Gabrielle N da Silva
- Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514 - Building 3, Porto Alegre, CEP 90050-170, RS, Brazil
| | - Adriana M Zago
- Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514 - Building 3, Porto Alegre, CEP 90050-170, RS, Brazil
| | - Luiz Felipe C Koenig
- Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514 - Building 3, Porto Alegre, CEP 90050-170, RS, Brazil
| | - Marilda C Fernandes
- Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Sarmento Leite, 245, Room 514 - Building 3, Porto Alegre, CEP 90050-170, RS, Brazil.
| |
Collapse
|
11
|
Wu SH, Ma J, Peng HE, Zheng RH, Zhang Z, Yang R, Wang L, Liu J, Mu J, Liu M, Li ZQ, Gong ZL, Shao JY, Zhong YW. Ratiometric Imaging Detection of Amyloid-β Fibrils by a Dual-Emissive Tris-Heteroleptic Ruthenium Complex. Inorg Chem 2024; 63:17983-17992. [PMID: 39287976 DOI: 10.1021/acs.inorgchem.4c02515] [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: 09/19/2024]
Abstract
Two dual fluorescent/phosphorescent tris-heteroleptic mononuclear Ru(ΙΙ) complexes (2 and 3) were designed and applied in amyloid-β (Aβ) sensing. These complexes have a general formula of [Ru(phen)(dppz)(L)](PF6)2, where L is (2-pyrazinyl)(2-pyridyl)(methyl)amine (H-L) with different substituents (-OMe for 2, -H for 3), phen is 1,10-phenanthroline, and dppz is dipyridophenazine, respectively. Compared with the previously reported ratiometric probe 1 with a di(pyrid-2-yl)(methyl)amine ligand, complex 2 can be employed for not only ratiometric emissive detection of Aβ aggregation but also ratiometric imaging detection of Aβ fibrils. In ratiometric emissive detection, as the incubation time of the Aβ sample (Aβ40 and Aβ42) was prolonged, a new phosphorescence emission band appeared with gradual enhancement of the emission intensity, while the fluorescence emission was basically unchanged, which could be treated as an intrinsic internal reference signal. In comparison, a larger ratiometric photoluminescence enhancement (I640/I440) was observed for Aβ40 aggregation with respect to Aβ42. In ratiometric imaging detection, the imaging signals obtained from the phosphorescence emission are much brighter than the fluorescence emission in both Aβ40 and Aβ42 fibrils. As indicated by molecular docking results, stronger interactions were found between complex 2 with Aβ40 fibrils, which included π/π, π/C-H, and π/H interactions between bidentate ligands dppz and phen with amino acid residues. Moreover, computational calculations were carried out to assist the interpretation of these experimental findings.
Collapse
Affiliation(s)
- Si-Hai Wu
- School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China
| | - Junjie Ma
- School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China
| | - Hui-E Peng
- School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China
| | - Ren-Hui Zheng
- School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China
| | - Zhe Zhang
- School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China
| | - Rong Yang
- School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China
| | - Lianhui Wang
- School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China
| | - Jieqing Liu
- School of Medicine, Huaqiao University, Quanzhou, Fujian 362021, China
| | - Jiao Mu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Meirong Liu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhong-Qiu Li
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhong-Liang Gong
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Jiang-Yang Shao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yu-Wu Zhong
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
12
|
Zedde M, Pascarella R. The Cerebrovascular Side of Plasticity: Microvascular Architecture across Health and Neurodegenerative and Vascular Diseases. Brain Sci 2024; 14:983. [PMID: 39451997 PMCID: PMC11506257 DOI: 10.3390/brainsci14100983] [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: 09/03/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
The delivery of nutrients to the brain is provided by a 600 km network of capillaries and microvessels. Indeed, the brain is highly energy demanding and, among a total amount of 100 billion neurons, each neuron is located just 10-20 μm from a capillary. This vascular network also forms part of the blood-brain barrier (BBB), which maintains the brain's stable environment by regulating chemical balance, immune cell transport, and blocking toxins. Typically, brain microvascular endothelial cells (BMECs) have low turnover, indicating a stable cerebrovascular structure. However, this structure can adapt significantly due to development, aging, injury, or disease. Temporary neural activity changes are managed by the expansion or contraction of arterioles and capillaries. Hypoxia leads to significant remodeling of the cerebrovascular architecture and pathological changes have been documented in aging and in vascular and neurodegenerative conditions. These changes often involve BMEC proliferation and the remodeling of capillary segments, often linked with local neuronal changes and cognitive function. Cerebrovascular plasticity, especially in arterioles, capillaries, and venules, varies over different time scales in development, health, aging, and diseases. Rapid changes in cerebral blood flow (CBF) occur within seconds due to increased neural activity. Prolonged changes in vascular structure, influenced by consistent environmental factors, take weeks. Development and aging bring changes over months to years, with aging-associated plasticity often improved by exercise. Injuries cause rapid damage but can be repaired over weeks to months, while neurodegenerative diseases cause slow, varied changes over months to years. In addition, if animal models may provide useful and dynamic in vivo information about vascular plasticity, humans are more complex to investigate and the hypothesis of glymphatic system together with Magnetic Resonance Imaging (MRI) techniques could provide useful clues in the future.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Rosario Pascarella
- Neuroradiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy;
| |
Collapse
|
13
|
Kolobova E, Petrushanko I, Mitkevich V, Makarov AA, Grigorova IL. β-Amyloids and Immune Responses Associated with Alzheimer's Disease. Cells 2024; 13:1624. [PMID: 39404388 PMCID: PMC11475064 DOI: 10.3390/cells13191624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is associated with the accumulation of β-amyloids (Aβs) and the formation of Aβ plaques in the brain. Various structural forms and isoforms of Aβs that have variable propensities for oligomerization and toxicity and may differentially affect the development of AD have been identified. In addition, there is evidence that β-amyloids are engaged in complex interactions with the innate and adaptive immune systems, both of which may also play a role in the regulation of AD onset and progression. In this review, we discuss what is currently known about the intricate interplay between β-amyloids and the immune response to Aβs with a more in-depth focus on the possible roles of B cells in the pathogenesis of AD.
Collapse
Affiliation(s)
- Elizaveta Kolobova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Irina Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
| | - Vladimir Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
| | - Irina L Grigorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (E.K.); (I.P.); (V.M.); (A.A.M.)
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| |
Collapse
|
14
|
Li Y, Awasthi S, Bryan L, Ehrlich RS, Tonali N, Balog S, Yang J, Sewald N, Mayer M. Fluorescence-Based Monitoring of Early-Stage Aggregation of Amyloid-β, Amylin Peptide, Tau, and α-Synuclein Proteins. ACS Chem Neurosci 2024; 15:3113-3123. [PMID: 39150403 PMCID: PMC11378287 DOI: 10.1021/acschemneuro.4c00097] [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: 02/12/2024] [Revised: 07/02/2024] [Accepted: 07/25/2024] [Indexed: 08/17/2024] Open
Abstract
Early-stage aggregates of amyloid-forming proteins, specifically soluble oligomers, are implicated in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Protein aggregation is typically monitored by fluorescence using the amyloid-binding fluorophore thioflavin T (ThT). Thioflavin T interacts, however, preferentially with fibrillar amyloid structures rather than with soluble, early-stage aggregates. In contrast, the two fluorophores, aminonaphthalene 2-cyanoacrylate-spiropyran (AN-SP) and triazole-containing boron-dipyrromethene (taBODIPY), were reported to bind preferentially to early-stage aggregates of amyloidogenic proteins. The present study compares ThT with AN-SP and taBODIPY with regard to their ability to monitor early stages of aggregation of four different amyloid-forming proteins, including amyloid-β (Aβ), tau protein, amylin, and α-synuclein. The results show that the three fluorophores vary in their suitability to monitor the early aggregation of different amyloid-forming proteins. For instance, in the presence of Aβ and amylin, the fluorescence intensity of AN-SP increased at an earlier stage of aggregation than the fluorescence of ThT, albeit with only a small fluorescence increase in the case of AN-SP. In contrast, in the presence of tau and amylin, the fluorescence intensity of taBODIPY increased at an earlier stage of aggregation than the fluorescence of ThT. Finally, α-synuclein aggregation could only be monitored by ThT fluorescence; neither AN-SP nor taBODIPY showed a significant increase in fluorescence over the course of aggregation of α-synuclein. These results demonstrate the ability of AN-SP and taBODIPY to monitor the formation of early-stage aggregates from specific amyloid-forming proteins at an early stage of aggregation, although moderate increases in fluorescence intensity, relatively large uncertainties in fluorescence values, and limited solubility of both fluorophores limit their usefulness for some amyloid proteins. The capability to monitor early aggregation of some amyloid proteins, such as amylin, might accelerate the discovery of aggregation inhibitors to minimize the formation of toxic oligomeric species for potential therapeutic use.
Collapse
Affiliation(s)
- Yuanjie Li
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| | - Saurabh Awasthi
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
- Department
of Biotechnology, National Institute of
Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow, Uttar Pradesh 226002, India
| | - Louise Bryan
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| | - Rachel S. Ehrlich
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093-0358, United States
| | - Nicolo Tonali
- CNRS,
BioCIS, Bâtiment Henri Moissan, Université
Paris-Saclay, 17 Av. des Sciences, Orsay 91400, France
| | - Sandor Balog
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| | - Jerry Yang
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093-0358, United States
| | - Norbert Sewald
- Bielefeld
University, Department of Chemistry P.O. Box 100131, Bielefeld 33501, Germany
| | - Michael Mayer
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| |
Collapse
|
15
|
Zhang C, Jia Q, Zhu L, Hou J, Wang X, Li D, Zhang J, Zhang Y, Yang S, Tu Z, Yan X, Yang W, Li S, Li X, Yin P. Suppressing UBE2N ameliorates Alzheimer's disease pathology through the clearance of amyloid beta. Alzheimers Dement 2024; 20:6287-6304. [PMID: 39015037 PMCID: PMC11497675 DOI: 10.1002/alz.14122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/28/2024] [Accepted: 06/13/2024] [Indexed: 07/18/2024]
Abstract
INTRODUCTION Aging is one of the risk factors for the early onset of Alzheimer's disease (AD). We previously discovered that the age-dependent increase in Ubiquitin Conjugating Enzyme E2 N (UBE2N) plays a role in the accumulation of misfolded proteins through K63 ubiquitination, which has been linked to AD pathogenesis. However, the impact of UBE2N on amyloid pathology and clearance has remained unknown. RESULTS We observed the elevated UBE2N during the amyloid beta (Aβ) generation in the brains of 5×FAD, APP/PS1 mice, and patients with AD, in comparison to healthy individuals. UBE2N overexpression exacerbated amyloid deposition in 5×FAD mice and senescent monkeys, whereas knocking down UBE2N via CRISPR/Cas9 reduced Aβ generation and cognitive deficiency. Moreover, pharmacological inhibition of UBE2N ameliorated Aβ pathology and subsequent transcript defects in 5×FAD mice. DISCUSSION We have discovered that age-dependent expression of UBE2N is a critical regulator of AD pathology. Our findings suggest that UBE2N could serve as a potential pharmacological target for the advancement of AD therapeutics. HIGHLIGHTS Ubiquitin Conjugating Enzyme E2 N (UBE2N) level was elevated during amyloid beta (Aβ) deposition in AD mouse and patients' brains. UBE2N exacerbated Aβ generation in the AD mouse and senescent monkey. Drug inhibition of UBE2N ameliorated Aβ pathology and cognitive deficiency.
Collapse
Affiliation(s)
- Chen Zhang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Qingqing Jia
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Longhong Zhu
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Junqi Hou
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Xiang Wang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Dandan Li
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Jiawei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Yiran Zhang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Su Yang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Zhuchi Tu
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Xiao‐Xin Yan
- Department of Anatomy and NeurobiologyXiangya School of MedicineCentral South UniversityChangshaChina
| | - Weili Yang
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Shihua Li
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Xiao‐Jiang Li
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Peng Yin
- State Key Laboratory of Bioactive Molecules and Druggability AssessmentGuangdong Key Laboratory of Non‐human Primate ResearchGuangdong‐Hongkong‐Macau Institute of CNS RegenerationJinan UniversityGuangzhouChina
| |
Collapse
|
16
|
Kolesnikova TO, Demin KA, Costa FV, de Abreu MS, Kalueff AV. Zebrafish models for studying cognitive enhancers. Neurosci Biobehav Rev 2024; 164:105797. [PMID: 38971515 DOI: 10.1016/j.neubiorev.2024.105797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/16/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Cognitive decline is commonly seen both in normal aging and in neurodegenerative and neuropsychiatric diseases. Various experimental animal models represent a valuable tool to study brain cognitive processes and their deficits. Equally important is the search for novel drugs to treat cognitive deficits and improve cognitions. Complementing rodent and clinical findings, studies utilizing zebrafish (Danio rerio) are rapidly gaining popularity in translational cognitive research and neuroactive drug screening. Here, we discuss the value of zebrafish models and assays for screening nootropic (cognitive enhancer) drugs and the discovery of novel nootropics. We also discuss the existing challenges, and outline future directions of research in this field.
Collapse
Affiliation(s)
| | - Konstantin A Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Fabiano V Costa
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; West Caspian University, Baku, Azerbaijan.
| | - Allan V Kalueff
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Suzhou Key Laboratory on Neurobiology and Cell Signaling, Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China.
| |
Collapse
|
17
|
Arafi P, Devkota S, Maesako M, Wolfe MS. Alzheimer-mutant γ-secretase complexes stall amyloid β-peptide production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610520. [PMID: 39257787 PMCID: PMC11383658 DOI: 10.1101/2024.08.30.610520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Missense mutations in the amyloid precursor protein (APP) and presenilin-1 (PSEN1) cause early-onset familial Alzheimer's disease (FAD) and alter proteolytic production of secreted 38-to-43-residue amyloid β-peptides (Aβ) by the PSEN1-containing γ-secretase complex, ostensibly supporting the amyloid hypothesis of pathogenesis. However, proteolysis of APP substrate by γ-secretase is processive, involving initial endoproteolysis to produce long Aβ peptides of 48 or 49 residues followed by carboxypeptidase trimming in mostly tripeptide increments. We recently reported evidence that FAD mutations in APP and PSEN1 cause deficiencies in early steps in processive proteolysis of APP substrate C99 and that this results from stalled γ-secretase enzyme-substrate and/or enzyme-intermediate complexes. These stalled complexes triggered synaptic degeneration in a C. elegans model of FAD independently of Aβ production. Here we conducted full quantitative analysis of all proteolytic events on APP substrate by γ-secretase with six additional PSEN1 FAD mutations and found that all six are deficient in multiple processing steps. However, only one of these (F386S) was deficient in certain trimming steps but not in endoproteolysis. Fluorescence lifetime imaging microscopy in intact cells revealed that all six PSEN1 FAD mutations lead to stalled γ-secretase enzyme-substrate/intermediate complexes. The F386S mutation, however, does so only in Aβ-rich regions of the cells, not in C99-rich regions, consistent with the deficiencies of this mutant enzyme only in trimming of Aβ intermediates. These findings provide further evidence that FAD mutations lead stalled and stabilized γ-secretase enzyme-substrate and/or enzyme-intermediate complexes and are consistent with the stalled process rather than the products of γ-secretase proteolysis as the pathogenic trigger.
Collapse
Affiliation(s)
- Parnian Arafi
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| | - Sujan Devkota
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| | - Masato Maesako
- Alzheimer Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
18
|
Terpstra K, Huang Y, Na H, Sun L, Gutierrez C, Yu Z, Mirica LM. 2-Phenylbenzothiazolyl iridium complexes as inhibitors and probes of amyloid β aggregation. Dalton Trans 2024; 53:14258-14264. [PMID: 39129539 PMCID: PMC11445708 DOI: 10.1039/d4dt01691b] [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] [Indexed: 08/13/2024]
Abstract
The aggregation of amyloid β (Aβ) peptides is a significant hallmark of Alzheimer's disease (AD), and the detection of Aβ aggregates and the inhibition of their formation are important for the diagnosis and treatment of AD, respectively. Herein, we report a series of benzothiazole-based Ir(III) complexes HN-1 to HN-8 that exhibit appreciable inhibition of Aβ aggregation in vitro and in living cells. These Ir(III) complexes can induce a significant fluorescence increase when binding to Aβ fibrils and Aβ oligomers, while their measured log D values suggest these compounds could have enhanced blood-brain barrier (BBB) permeability. In vivo studies show that HN-1, HN-2, HN-3, and HN-8 successfully penetrate the BBB and stain the amyloid plaques in AD mouse brains after a 10-day treatment, suggesting that these Ir(III) complexes could act as lead compounds for AD therapeutic and diagnostic agent development.
Collapse
Affiliation(s)
- Karna Terpstra
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, USA.
| | - Yiran Huang
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, USA.
| | - Hanah Na
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, USA.
| | - Liang Sun
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, USA.
| | - Citlali Gutierrez
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, USA.
| | - Zhengxin Yu
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, USA.
| | - Liviu M Mirica
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, USA.
| |
Collapse
|
19
|
Xu CK, Meisl G, Andrzejewska EA, Krainer G, Dear AJ, Castellana-Cruz M, Turi S, Edu IA, Vivacqua G, Jacquat RPB, Arter WE, Spillantini MG, Vendruscolo M, Linse S, Knowles TPJ. α-Synuclein oligomers form by secondary nucleation. Nat Commun 2024; 15:7083. [PMID: 39153989 PMCID: PMC11330488 DOI: 10.1038/s41467-024-50692-4] [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: 08/27/2023] [Accepted: 07/19/2024] [Indexed: 08/19/2024] Open
Abstract
Oligomeric species arising during the aggregation of α-synuclein are implicated as a major source of toxicity in Parkinson's disease, and thus a major potential drug target. However, both their mechanism of formation and role in aggregation are largely unresolved. Here we show that, at physiological pH and in the absence of lipid membranes, α-synuclein aggregates form by secondary nucleation, rather than simple primary nucleation, and that this process is enhanced by agitation. Moreover, using a combination of single molecule and bulk level techniques, we identify secondary nucleation on the surfaces of existing fibrils, rather than formation directly from monomers, as the dominant source of oligomers. Our results highlight secondary nucleation as not only the key source of oligomers, but also the main mechanism of aggregate formation, and show that these processes take place under conditions which recapitulate the neutral pH and ionic strength of the cytosol.
Collapse
Affiliation(s)
- Catherine K Xu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- Max Planck Institute for the Science of Light, Erlangen, Germany
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Ewa A Andrzejewska
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Georg Krainer
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- Institute of Molecular Biosciences (IMB), University of Graz, Graz, Austria
| | - Alexander J Dear
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Marta Castellana-Cruz
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Soma Turi
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Irina A Edu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Giorgio Vivacqua
- Integrated Research Center (PRAAB), Campus Biomedico University of Rome, Rome, Italy
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Raphaël P B Jacquat
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - William E Arter
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | | | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Sara Linse
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
- Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
Sheikh AM, Tabassum S, Yano S, Abdullah FB, Wang R, Ikeue T, Nagai A. A Cationic Zn-Phthalocyanine Turns Alzheimer's Amyloid β Aggregates into Non-Toxic Oligomers and Inhibits Neurotoxicity in Culture. Int J Mol Sci 2024; 25:8931. [PMID: 39201616 PMCID: PMC11354870 DOI: 10.3390/ijms25168931] [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: 07/17/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Amyloid β peptide (Aβ) aggregation and deposition are considered the main causes of Alzheimer's disease. In a previous study, we demonstrated that anionic Zn-phthalocyanine (ZnPc) can interact with the Aβ peptide and inhibit the fibril-formation process. However, due to the inability of anionic ZnPc to cross the intact blood-brain barrier, we decided to explore the interaction of cationic methylated Zn-phthalocyanine (cZnPc) with the peptide. Using a ThT fluorescence assay, we observed that cZnPc dose-dependently and time-dependently inhibited Aβ1-42 fibril levels under in vitro fibril-formation conditions. Electron microscopy revealed that it caused Aβ1-42 peptides to form small aggregates. Western blotting and dot immunoblot oligomer experiments demonstrated that cZnPc increased rather than decreased the levels of oligomers from the very early stages of incubation. A binding assay confirmed that cZnPc could bind with the peptide. Docking simulations indicated that the oligomer species of Aβ1-42 had a higher ability to interact with cZnPc. ANS fluorescence assay results indicated that cZnPc did not affect the hydrophobicity of the peptide. However, cZnPc significantly increased intrinsic tyrosine fluorescence of the peptide after 8 h of incubation in fibril-formation conditions. Importantly, cell culture experiments demonstrated that cZnPc did not exhibit any toxicity up to a concentration of 10 µM. Instead, it protected a neuronal cell line from Aβ1-42-induced toxicity. Thus, our results suggest that cZnPc can affect the aggregation process of Aβ1-42, rendering it non-toxic, which could be crucial for the therapy of Alzheimer's disease.
Collapse
Affiliation(s)
- Abdullah Md. Sheikh
- Department of Laboratory Medicine, Shimane University Faculty of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.T.); (S.Y.); (A.N.)
| | - Shatera Tabassum
- Department of Laboratory Medicine, Shimane University Faculty of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.T.); (S.Y.); (A.N.)
| | - Shozo Yano
- Department of Laboratory Medicine, Shimane University Faculty of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.T.); (S.Y.); (A.N.)
| | - Fatema Binte Abdullah
- Department of Neurology, Shimane University Faculty of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan; (F.B.A.); (R.W.)
| | - Ruochen Wang
- Department of Neurology, Shimane University Faculty of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan; (F.B.A.); (R.W.)
| | - Takahisa Ikeue
- Department of Chemistry, Graduate School of Science and Engineering, Shimane University, 1060 Nishikawatsu, Matsue 690-8504, Japan;
| | - Atsushi Nagai
- Department of Laboratory Medicine, Shimane University Faculty of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan; (S.T.); (S.Y.); (A.N.)
- Department of Neurology, Shimane University Faculty of Medicine, 89-1 Enya Cho, Izumo 693-8501, Japan; (F.B.A.); (R.W.)
| |
Collapse
|
21
|
Velázquez-Delgado C, Hernández-Ortiz E, Landa-Navarro L, Tapia-Rodríguez M, Moreno-Castilla P, Bermúdez-Rattoni F. Repeated exposure to novelty promotes resilience against the amyloid-beta effect through dopaminergic stimulation. Psychopharmacology (Berl) 2024:10.1007/s00213-024-06650-5. [PMID: 39145803 DOI: 10.1007/s00213-024-06650-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/07/2024] [Indexed: 08/16/2024]
Abstract
RATIONALE The accumulation of beta-amyloid peptide (Aβ) in the forebrain leads to cognitive dysfunction and neurodegeneration in Alzheimer's disease. Studies have shown that individuals with a consistently cognitively active lifestyle are less vulnerable to Aβ toxicity. Recent research has demonstrated that intrahippocampal Aβ can impact catecholaminergic release and spatial memory. Interestingly, exposure to novelty stimuli has been found to stimulate the release of catecholamines in the hippocampus. However, it remains uncertain whether repeated enhancing catecholamine activity can effectively alleviate cognitive impairment in individuals with Alzheimer's disease. OBJECTIVES Our primary aim was to investigate whether repeated exposure to novelty could enable cognitive resilience against Aβ. This protection could be achieved by modulating catecholaminergic activity within the hippocampus. METHODS To investigate this hypothesis, we subjected mice to three different conditions-standard housing (SH), repeated novelty (Nov), or daily social interaction (Soc) for one month. We then infused saline solution (SS) or Aβ (Aβ1-42) oligomers intrahippocampally and measured spatial memory retrieval in a Morris Water Maze (MWM). Stereological analysis and extracellular baseline dopamine levels using in vivo microdialysis were assessed in independent groups of mice. RESULTS The mice that received Aβ1-42 intrahippocampal infusions and remained in SH or Soc conditions showed impaired spatial memory retrieval. In contrast, animals subjected to the Nov protocol demonstrated remarkable resilience, showing strong spatial memory expression even after Aβ1-42 intrahippocampal infusion. The stereological analysis indicated that the Aβ1-42 infusion reduced the tyrosine hydroxylase axonal length in SH or Soc mice compared to the Nov group. Accordingly, the hippocampal extracellular dopamine levels increased significantly in the Nov groups. CONCLUSIONS These compelling results demonstrate the potential for repeated novelty exposure to strengthen the dopaminergic system and mitigate the toxic effects of Aβ1-42. They also highlight new and promising therapeutic avenues for treating and preventing AD, especially in its early stages.
Collapse
Affiliation(s)
- Cintia Velázquez-Delgado
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Eduardo Hernández-Ortiz
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Lucia Landa-Navarro
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Miguel Tapia-Rodríguez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Perla Moreno-Castilla
- Laboratory of Cognitive Resilience, Center of Aging Research (CIE), Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV, Mexico City, Mexico.
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
22
|
Honda K, Takahashi H, Hata S, Abe R, Saito T, Saido TC, Taru H, Sobu Y, Ando K, Yamamoto T, Suzuki T. Suppression of the amyloidogenic metabolism of APP and the accumulation of Aβ by alcadein α in the brain during aging. Sci Rep 2024; 14:18471. [PMID: 39122814 PMCID: PMC11316129 DOI: 10.1038/s41598-024-69400-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
Generation and accumulation of amyloid-β (Aβ) protein in the brain are the primary causes of Alzheimer's disease (AD). Alcadeins (Alcs composed of Alcα, Alcβ and Alcγ family) are a neuronal membrane protein that is subject to proteolytic processing, as is Aβ protein precursor (APP), by APP secretases. Previous observations suggest that Alcs are involved in the pathophysiology of Alzheimer's disease (AD). Here, we generated new mouse AppNL-F (APP-KI) lines with either Alcα- or Alcβ-deficient background and analyzed APP processing and Aβ accumulation through the aging process. The Alcα-deficient APP-KI (APP-KI/Alcα-KO) mice enhanced brain Aβ accumulation along with increased amyloidogenic β-site cleavage of APP through the aging process whereas Alcβ-deficient APP-KI (APP-KI/Alcβ-KO) mice neither affected APP metabolism nor Aβ accumulation at any age. More colocalization of APP and BACE1 was observed in the endolysosomal pathway in neurons of APP-KI/Alcα-KO mice compared to APP-KI and APP-KI/Alcβ-KO mice. These results indicate that Alcα plays an important role in the neuroprotective function by suppressing the amyloidogenic cleavage of APP by BACE1 in the brain, which is distinct from the neuroprotective function of Alcβ, in which p3-Alcβ peptides derived from Alcβ restores the viability in neurons impaired by toxic Aβ.
Collapse
Affiliation(s)
- Keiko Honda
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Hiroo Takahashi
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, 761-0793, Japan
| | - Saori Hata
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo, 062-8517, Japan
| | - Ruriko Abe
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo, 062-8517, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science Institute, Wako, 351-0198, Japan
| | - Hidenori Taru
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Yuriko Sobu
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
- Laboratory of Neuronal Regeneration, Graduate School of Brain Science, Doshisha University, Kyotanabe, 610-0394, Japan
| | - Kanae Ando
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Miki-cho, 761-0793, Japan.
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
- Advanced Prevention and Research Laboratory for Dementia, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan.
| |
Collapse
|
23
|
Vieira TCRG, Barros CA, Domingues R, Outeiro TF. PrP meets alpha-synuclein: Molecular mechanisms and implications for disease. J Neurochem 2024; 168:1625-1639. [PMID: 37855859 DOI: 10.1111/jnc.15992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/19/2023] [Accepted: 10/01/2023] [Indexed: 10/20/2023]
Abstract
The discovery of prions has challenged dogmas and has revolutionized our understanding of protein-misfolding diseases. The concept of self-propagation via protein conformational changes, originally discovered for the prion protein (PrP), also applies to other proteins that exhibit similar behavior, such as alpha-synuclein (aSyn), a central player in Parkinson's disease and in other synucleinopathies. aSyn pathology appears to spread from one cell to another during disease progression, and involves the misfolding and aggregation of aSyn. How the transfer of aSyn between cells occurs is still being studied, but one important hypothesis involves receptor-mediated transport. Interestingly, recent studies indicate that the cellular prion protein (PrPC) may play a crucial role in this process. PrPC has been shown to act as a receptor/sensor for protein aggregates in different neurodegenerative disorders, including Alzheimer's disease and amyotrophic lateral sclerosis. Here, we provide a comprehensive overview of the current state of knowledge regarding the interaction between aSyn and PrPC and discuss its role in synucleinopathies. We examine the properties of PrP and aSyn, including their structure, function, and aggregation. Additionally, we discuss the current understanding of PrPC's role as a receptor/sensor for aSyn aggregates and identify remaining unanswered questions in this area of research. Ultimately, we posit that exploring the interaction between aSyn and PrPC may offer potential treatment options for synucleinopathies.
Collapse
Affiliation(s)
- Tuane C R G Vieira
- Institute of Medical Biochemistry Leopoldo de Meis and National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline A Barros
- Institute of Medical Biochemistry Leopoldo de Meis and National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato Domingues
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| |
Collapse
|
24
|
Wang L, Tang Z, Li B, Peng Y, Yang X, Xiao Y, Ni R, Qi XL. Myricetin ameliorates cognitive impairment in 3×Tg Alzheimer's disease mice by regulating oxidative stress and tau hyperphosphorylation. Biomed Pharmacother 2024; 177:116963. [PMID: 38889642 DOI: 10.1016/j.biopha.2024.116963] [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: 04/22/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Alzheimer's disease is characterized by abnormal β-amyloid (Aβ) plaque accumulation, tau hyperphosphorylation, reactive oxidative stress, mitochondrial dysfunction and synaptic loss. Myricetin, a dietary flavonoid, has been shown to exert neuroprotective effects in vitro and in vivo. Here, we aimed to elucidate the mechanism and pathways involved in the protective effect of myricetin. METHODS The effect of myricetin was assessed on Aβ42 oligomer-treated neuronal SH-SY5Y cells and in 3×Tg mice. Behavioral tests were performed to assess the cognitive effects of myricetin (14 days, ip) in 3×Tg mice. The levels of beta-amyloid precursor protein (APP), synaptic and mitochondrial proteins, glycogen synthase kinase3β (GSK3β) and extracellular regulated kinase (ERK) 2 were assessed via Western blotting. Flow cytometry assays, immunofluorescence staining, and transmission electron microscopy were used to assess mitochondrial dysfunction and reactive oxidative stress. RESULTS We found that, compared with control treatment, myricetin treatment improved spatial cognition and learning and memory in 3×Tg mice. Myricetin ameliorated tau phosphorylation and the reduction in pre- and postsynaptic proteins in Aβ42 oligomer-treated neuronal SH-SY5Y cells and in 3×Tg mice. In addition, myricetin reduced reactive oxygen species generation, lipid peroxidation, and DNA oxidation, and rescued mitochondrial dysfunction via the associated GSK3β and ERK 2 signalling pathways. CONCLUSIONS This study provides new insight into the neuroprotective mechanism of myricetin in vitro in cell culture and in vivo in a mouse model of Alzheimer's disease.
Collapse
Affiliation(s)
- Li Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Zhi Tang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Bo Li
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Yaqian Peng
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Xi Yang
- Guiyang Healthcare Vocational University, Guizhou ERC for Medical Resources & Healthcare Products (Guizhou Engineering Research Center for Medical Resources and Healthcare Products), Guiyang, Guizhou, China
| | - Yan Xiao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland; Institute for Biomedical Engineering, ETH Zurich & University of Zurich, Zurich, Switzerland.
| | - Xiao-Lan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Constructed by the Province and Ministry, Guiyang, China.
| |
Collapse
|
25
|
Leimu L, Holm P, Gąciarz A, Haavisto O, Prince S, Pesonen U, Huovinen T, Lamminmäki U. Epitope-specific antibody fragments block aggregation of AGelD187N, an aberrant peptide in gelsolin amyloidosis. J Biol Chem 2024; 300:107507. [PMID: 38944121 PMCID: PMC11298591 DOI: 10.1016/j.jbc.2024.107507] [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: 04/26/2024] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024] Open
Abstract
Aggregation of aberrant fragment of plasma gelsolin, AGelD187N, is a crucial event underlying the pathophysiology of Finnish gelsolin amyloidosis, an inherited form of systemic amyloidosis. The amyloidogenic gelsolin fragment AGelD187N does not play any physiological role in the body, unlike most aggregating proteins related to other protein misfolding diseases. However, no therapeutic agents that specifically and effectively target and neutralize AGelD187N exist. We used phage display technology to identify novel single-chain variable fragments that bind to different epitopes in the monomeric AGelD187N that were further maturated by variable domain shuffling and converted to antigen-binding fragment (Fab) antibodies. The generated antibody fragments had nanomolar binding affinity for full-length AGelD187N, as evaluated by biolayer interferometry. Importantly, all four Fabs selected for functional studies efficiently inhibited the amyloid formation of full-length AGelD187N as examined by thioflavin fluorescence assay and transmission electron microscopy. Two Fabs, neither of which bound to the previously proposed fibril-forming region of AGelD187N, completely blocked the amyloid formation of AGelD187N. Moreover, no small soluble aggregates, which are considered pathogenic species in protein misfolding diseases, were formed after successful inhibition of amyloid formation by the most promising aggregation inhibitor, as investigated by size-exclusion chromatography combined with multiangle light scattering. We conclude that all regions of the full-length AGelD187N are important in modulating its assembly into fibrils and that the discovered epitope-specific anti-AGelD187N antibody fragments provide a promising starting point for a disease-modifying therapy for gelsolin amyloidosis, which is currently lacking.
Collapse
Affiliation(s)
- Laura Leimu
- R&D, Orion Pharma, Orion Corporation, Turku, Finland; Faculty of Medicine, Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Patrik Holm
- R&D, Orion Pharma, Orion Corporation, Turku, Finland; Department of Life Technologies, University of Turku, Turku, Finland; Organon R&D Finland, Turku, Finland
| | - Anna Gąciarz
- R&D, Orion Pharma, Orion Corporation, Turku, Finland; Mobidiag, A Hologic Company, Espoo, Finland
| | - Oskar Haavisto
- Department of Life Technologies, University of Turku, Turku, Finland
| | - Stuart Prince
- R&D, Orion Pharma, Orion Corporation, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Ullamari Pesonen
- Faculty of Medicine, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tuomas Huovinen
- Department of Life Technologies, University of Turku, Turku, Finland
| | - Urpo Lamminmäki
- Department of Life Technologies, University of Turku, Turku, Finland.
| |
Collapse
|
26
|
Moraca F, Vespoli I, Mastroianni D, Piscopo V, Gaglione R, Arciello A, De Nisco M, Pacifico S, Catalanotti B, Pedatella S. Synthesis, biological evaluation and metadynamics simulations of novel N-methyl β-sheet breaker peptides as inhibitors of Alzheimer's β-amyloid fibrillogenesis. RSC Med Chem 2024; 15:2286-2299. [PMID: 39026638 PMCID: PMC11253850 DOI: 10.1039/d4md00057a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/07/2024] [Indexed: 07/20/2024] Open
Abstract
Several scientific evidences report that a central role in the pathogenesis of Alzheimer's disease is played by the deposition of insoluble aggregates of β-amyloid proteins in the brain. Because Aβ is self-assembling, one possible design strategy is to inhibit the aggregation of Aβ peptides using short peptide fragments homologous to the full-length wild-type Aβ protein. In the past years, several studies have reported on the synthesis of some short synthetic peptides called β-sheet breaker peptides (BSBPs). Herein, we present the synthesis of novel (cell-permeable) N-methyl BSBPs, designed based on literature information on the structural key features of BSBPs. Three-dimensional GRID-based pharmacophore peptide screening combined with PT-WTE metadynamics was performed to support the results of the design and microwave-assisted synthesis of peptides 2 and 3 prepared and analyzed for their fibrillogenesis inhibition activity and cytotoxicity. An HR-MS-based cell metabolomic approach highlighted their cell permeability properties.
Collapse
Affiliation(s)
- Federica Moraca
- Department of Pharmacy, University of Napoli Federico II Via Domenico Montesano 49 I-80131 Napoli Italy
- Net4Science Academic Spin-Off, University "Magna Græcia" of Catanzaro Viale Europa 88100 Catanzaro Italy
| | - Ilaria Vespoli
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Flemingovo náměstí 542/2 CZ-16610 Prague Czech Republic
| | - Domenico Mastroianni
- Department of Chemical Sciences, University of Napoli Federico II Via Cintia 4 I-80126 Napoli Italy
| | - Vincenzo Piscopo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli" Viale Abramo Lincoln 5 I-81100 Caserta Italy
| | - Rosa Gaglione
- Department of Chemical Sciences, University of Napoli Federico II Via Cintia 4 I-80126 Napoli Italy
- Istituto Nazionale di Biostrutture e Biosistemi (INBB) Viale delle Medaglie d'Oro 305 I-80145 Roma Italy
| | - Angela Arciello
- Department of Chemical Sciences, University of Napoli Federico II Via Cintia 4 I-80126 Napoli Italy
- Istituto Nazionale di Biostrutture e Biosistemi (INBB) Viale delle Medaglie d'Oro 305 I-80145 Roma Italy
| | - Mauro De Nisco
- Department of Sciences, University of Basilicata Viale dell'Ateneo Lucano I-85100 Potenza Italy
| | - Severina Pacifico
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli" Viale Abramo Lincoln 5 I-81100 Caserta Italy
| | - Bruno Catalanotti
- Department of Pharmacy, University of Napoli Federico II Via Domenico Montesano 49 I-80131 Napoli Italy
| | - Silvana Pedatella
- Department of Chemical Sciences, University of Napoli Federico II Via Cintia 4 I-80126 Napoli Italy
| |
Collapse
|
27
|
González Díaz A, Cataldi R, Mannini B, Vendruscolo M. Preparation and Characterization of Zn(II)-Stabilized Aβ 42 Oligomers. ACS Chem Neurosci 2024; 15:2586-2599. [PMID: 38979921 PMCID: PMC11258685 DOI: 10.1021/acschemneuro.4c00084] [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: 02/06/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
Aβ oligomers are being investigated as cytotoxic agents in Alzheimer's disease (AD). Because of their transient nature and conformational heterogeneity, the relationship between the structure and activity of these oligomers is still poorly understood. Hence, methods for stabilizing Aβ oligomeric species relevant to AD are needed to uncover the structural determinants of their cytotoxicity. Here, we build on the observation that metal ions and metabolites have been shown to interact with Aβ, influencing its aggregation and stabilizing its oligomeric species. We thus developed a method that uses zinc ions, Zn(II), to stabilize oligomers produced by the 42-residue form of Aβ (Aβ42), which is dysregulated in AD. These Aβ42-Zn(II) oligomers are small in size, spanning the 10-30 nm range, stable at physiological temperature, and with a broad toxic profile in human neuroblastoma cells. These oligomers offer a tool to study the mechanisms of toxicity of Aβ oligomers in cellular and animal AD models.
Collapse
Affiliation(s)
- Alicia González Díaz
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Rodrigo Cataldi
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Benedetta Mannini
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
- Department
of Experimental and Clinical Biomedical Sciences Mario Serio, University
of Florence, 50134 Florence, Italy
| | - Michele Vendruscolo
- Centre
for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
28
|
Tang Y, Wang Y, Gao Z, Li J, Zhang L, Shi H, Dong J, Song S, Qian C. sAPPα Peptide Promotes Damaged Microglia to Clear Alzheimer's Amyloid-β via Restoring Mitochondrial Function. Chemistry 2024; 30:e202400870. [PMID: 38736169 DOI: 10.1002/chem.202400870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease with amyloid-β (Aβ) deposition as the main pathological feature. It's an important challenge to find new ways to clear Aβ from the brain. The soluble amyloid precursor protein α (sAPPα) is a neuroprotective protein and can attenuate neuronal damage, including toxic Aβ. However, the regulatory role of sAPPα in non-neuronal cells, such as microglia, is less reported and controversial. Here, we showed that sAPPα promoted the phagocytosis and degradation of Aβ in both normal and damaged microglia. Moreover, the function of damaged microglia was improved by the sAPPα through normalizing mitochondrial function. Furthermore, the results of molecular docking simulation showed that sAPPα had a good affinity with Aβ. We preliminarily reveal that sAPPα is similar to antibodies and can participate in the regulation of microglia phagocytosis and degradation of Aβ after binding to Aβ. sAPPα is expected to be a mild and safe peptide drug or drug carrier for AD.
Collapse
Affiliation(s)
- Yingqi Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Yangang Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Ziran Gao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Jiayi Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Lijia Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Haoting Shi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Jingwen Dong
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Shipeng Song
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Chenggen Qian
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| |
Collapse
|
29
|
Lima MP, Hornsby BD, Lim CS, Cheatham TE. Molecular Modeling of Single- and Double-Hydrocarbon-Stapled Coiled-Coil Inhibitors against Bcr-Abl: Toward a Treatment Strategy for CML. J Phys Chem B 2024; 128:6476-6491. [PMID: 38951498 PMCID: PMC11247501 DOI: 10.1021/acs.jpcb.4c02699] [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: 04/25/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/03/2024]
Abstract
The chimeric oncoprotein Bcr-Abl is the causative agent of virtually all chronic myeloid leukemias and a subset of acute lymphoblastic leukemias. As a result of the so-called Philadelphia chromosome translocation t(9;22), Bcr-Abl manifests as a constitutively active tyrosine kinase, which promotes leukemogenesis by activation of cell cycle signaling pathways. Constitutive and oncogenic activation is mediated by an N-terminal coiled-coil oligomerization domain in Bcr (Bcr-CC), presenting a therapeutic target for inhibition of Bcr-Abl activity toward the treatment of Bcr-Abl+ leukemias. Previously, we demonstrated that a rationally designed Bcr-CC mutant, CCmut3, exerts a dominant negative effect upon Bcr-Abl activity by preferential oligomerization with Bcr-CC. Moreover, we have shown that conjugation to a leukemia-specific cell-penetrating peptide (CPP-CCmut3) improves intracellular delivery and activity. However, our full-length CPP-CCmut3 construct (81 aa) is encumbered by an intrinsically high degree of conformational variability and susceptibility to proteolytic degradation relative to traditional small-molecule therapeutics. Here, we iterate a new generation of CCmut3 inhibitors against Bcr-CC-mediated Bcr-Abl assembly designed to address these constraints through incorporation of all-hydrocarbon staples spanning i and i + 7 positions in α-helix 2 (CPP-CCmut3-st). We utilize computational modeling and biomolecular simulation to evaluate single- and double-stapled CCmut3 candidates in silico for dynamics and binding energetics. We further model a truncated system characterized by the deletion of α-helix 1 and the flexible loop linker, which are known to impart high conformational variability. To study the impact of the N-terminal cyclic CPP toward model stability and inhibitor activity, we also model the full-length and truncated systems devoid of the CPP, with a cyclized CPP, and with an open-configuration CPP, for a total of six systems that comprise our library. From this library, we present lead-stapled peptide candidates to be synthesized and evaluated experimentally as our next iteration of inhibitors against Bcr-Abl.
Collapse
MESH Headings
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- Fusion Proteins, bcr-abl/chemistry
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Humans
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/metabolism
- Models, Molecular
- Molecular Dynamics Simulation
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Cell-Penetrating Peptides/chemistry
- Cell-Penetrating Peptides/pharmacology
- Cell-Penetrating Peptides/metabolism
Collapse
Affiliation(s)
- Maria
Carolina P. Lima
- Department
of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Braxten D. Hornsby
- Department
of Molecular Pharmaceutics, University of
Utah, Salt Lake City, Utah 84112, United States
| | - Carol S. Lim
- Department
of Molecular Pharmaceutics, University of
Utah, Salt Lake City, Utah 84112, United States
| | - Thomas E. Cheatham
- Department
of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
30
|
Mitra A, Naik L, Dhiman R, Sarkar N. Protonation-State Dependent Modulation of Hen Egg-White Lysozyme Fibrillation under the Influence of a Short Synthetic Peptide. J Phys Chem B 2024; 128:5995-6013. [PMID: 38875472 DOI: 10.1021/acs.jpcb.4c01578] [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: 06/16/2024]
Abstract
Under the influence of various conditions, misfolding of soluble proteins occurs, leading to the formation of toxic insoluble amyloids. The formation and deposition of such amyloids within the body are associated with detrimental biological consequences such as the onset of several amyloid-related diseases. Previously, we established a strategy for the rational design of peptide inhibitors against amyloid formation based on the amyloidogenic-prone region of the protein. In the current study, we have designed and identified an Asp-containing rationally designed hexapeptide (SqP4) as an excellent inhibitor of hen egg-white lysozyme (HEWL) amyloid progression in vitro. First, SqP4 showed strong affinity toward the native monomeric HEWL leading to the stabilization of the native form and restriction in the unfolding process of monomeric HEWL. Second, SqP4 was found to arrest the amyloidogenic misfolded structure of HEWL in a nonfibrillar monomer-like stage. We also observed the differential effect of the protonation state of the charged amino acid (Asp) within the peptide inhibitor on the amyloid formation of HEWL and explored the reason behind the observations. The findings of this study can be implemented in future strategies for the development of potent therapeutics against other amyloid-related diseases.
Collapse
Affiliation(s)
- Amit Mitra
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Lincoln Naik
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Nandini Sarkar
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| |
Collapse
|
31
|
Marques S, Kouba P, Legrand A, Sedlar J, Disson L, Planas-Iglesias J, Sanusi Z, Kunka A, Damborsky J, Pajdla T, Prokop Z, Mazurenko S, Sivic J, Bednar D. CoVAMPnet: Comparative Markov State Analysis for Studying Effects of Drug Candidates on Disordered Biomolecules. JACS AU 2024; 4:2228-2245. [PMID: 38938816 PMCID: PMC11200249 DOI: 10.1021/jacsau.4c00182] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/24/2024] [Accepted: 05/13/2024] [Indexed: 06/29/2024]
Abstract
Computational study of the effect of drug candidates on intrinsically disordered biomolecules is challenging due to their vast and complex conformational space. Here, we developed a comparative Markov state analysis (CoVAMPnet) framework to quantify changes in the conformational distribution and dynamics of a disordered biomolecule in the presence and absence of small organic drug candidate molecules. First, molecular dynamics trajectories are generated using enhanced sampling, in the presence and absence of small molecule drug candidates, and ensembles of soft Markov state models (MSMs) are learned for each system using unsupervised machine learning. Second, these ensembles of learned MSMs are aligned across different systems based on a solution to an optimal transport problem. Third, the directional importance of inter-residue distances for the assignment to different conformational states is assessed by a discriminative analysis of aggregated neural network gradients. This final step provides interpretability and biophysical context to the learned MSMs. We applied this novel computational framework to assess the effects of ongoing phase 3 therapeutics tramiprosate (TMP) and its metabolite 3-sulfopropanoic acid (SPA) on the disordered Aβ42 peptide involved in Alzheimer's disease. Based on adaptive sampling molecular dynamics and CoVAMPnet analysis, we observed that both TMP and SPA preserved more structured conformations of Aβ42 by interacting nonspecifically with charged residues. SPA impacted Aβ42 more than TMP, protecting α-helices and suppressing the formation of aggregation-prone β-strands. Experimental biophysical analyses showed only mild effects of TMP/SPA on Aβ42 and activity enhancement by the endogenous metabolization of TMP into SPA. Our data suggest that TMP/SPA may also target biomolecules other than Aβ peptides. The CoVAMPnet method is broadly applicable to study the effects of drug candidates on the conformational behavior of intrinsically disordered biomolecules.
Collapse
Affiliation(s)
- Sérgio
M. Marques
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, Brno 656
91, Czech Republic
| | - Petr Kouba
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- Czech
Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, Dejvice, Praha 6 160 00, Czech Republic
- Faculty
of Electrical Engineering, Czech Technical
University in Prague, Technicka 2, Dejvice, Praha 6 166 27, Czech Republic
| | - Anthony Legrand
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, Brno 656
91, Czech Republic
| | - Jiri Sedlar
- Czech
Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, Dejvice, Praha 6 160 00, Czech Republic
| | - Lucas Disson
- Czech
Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, Dejvice, Praha 6 160 00, Czech Republic
| | - Joan Planas-Iglesias
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, Brno 656
91, Czech Republic
| | - Zainab Sanusi
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, Brno 656
91, Czech Republic
| | - Antonin Kunka
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, Brno 656
91, Czech Republic
| | - Jiri Damborsky
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, Brno 656
91, Czech Republic
| | - Tomas Pajdla
- Czech
Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, Dejvice, Praha 6 160 00, Czech Republic
| | - Zbynek Prokop
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, Brno 656
91, Czech Republic
| | - Stanislav Mazurenko
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, Brno 656
91, Czech Republic
| | - Josef Sivic
- Czech
Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, Dejvice, Praha 6 160 00, Czech Republic
| | - David Bednar
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, Brno 656
91, Czech Republic
| |
Collapse
|
32
|
Kitamura A, Fujimoto A, Kawashima R, Lyu Y, Sasaki K, Hamada Y, Moriya K, Kurata A, Takahashi K, Brielmann R, Bott LC, Morimoto RI, Kinjo M. Hetero-oligomerization of TDP-43 carboxy-terminal fragments with cellular proteins contributes to proteotoxicity. Commun Biol 2024; 7:743. [PMID: 38902525 PMCID: PMC11190292 DOI: 10.1038/s42003-024-06410-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
Carboxy terminal fragments (CTFs) of TDP-43 contain an intrinsically disordered region (IDR) and form cytoplasmic condensates containing amyloid fibrils. Such condensates are toxic and associated with pathogenicity in amyotrophic lateral sclerosis. However, the molecular details of how the domain of TDP-43 CTFs leads to condensation and cytotoxicity remain elusive. Here, we show that truncated RNA/DNA-recognition motif (RRM) at the N-terminus of TDP-43 CTFs leads to the structural transition of the IDR, whereas the IDR itself of TDP-43 CTFs is difficult to assemble even if they are proximate intermolecularly. Hetero-oligomers of TDP-43 CTFs that have recruited other proteins are more toxic than homo-oligomers, implicating loss-of-function of the endogenous proteins by such oligomers is associated with cytotoxicity. Furthermore, such toxicity of TDP-43 CTFs was cell-nonautonomously affected in the nematodes. Therefore, misfolding and oligomeric characteristics of the truncated RRM at the N-terminus of TDP-43 CTFs define their condensation properties and toxicity.
Collapse
Affiliation(s)
- Akira Kitamura
- Laboratory of Cellular and Molecular Sciences, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan.
- PRIME, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, 100-0004, Japan.
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan.
| | - Ai Fujimoto
- Laboratory of Cellular and Molecular Sciences, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan
| | - Rei Kawashima
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan
| | - Yidan Lyu
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan
| | - Kotetsu Sasaki
- Laboratory of Cellular and Molecular Sciences, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan
| | - Yuta Hamada
- Laboratory of Cellular and Molecular Sciences, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan
| | - Kanami Moriya
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan
| | - Ayumi Kurata
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan
| | - Kazuho Takahashi
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan
| | - Reneé Brielmann
- Department of Molecular Bioscience, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, 60208, USA
| | - Laura C Bott
- Department of Molecular Bioscience, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, 60208, USA
| | - Richard I Morimoto
- Department of Molecular Bioscience, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, 60208, USA
| | - Masataka Kinjo
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo, 001-0021, Japan
| |
Collapse
|
33
|
Yin X, Zhou H, Cao T, Yang X, Meng F, Dai X, Wang Y, Li S, Zhai W, Yang Z, Chen N, Zhou R. Rational Design of Dual-Functionalized Gd@C 82 Nanoparticles to Relieve Neuronal Cytotoxicity in Alzheimer's Disease via Inhibition of Aβ Aggregation. ACS NANO 2024; 18:15416-15431. [PMID: 38840269 DOI: 10.1021/acsnano.3c08823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
The accumulation of amyloid-β (Aβ) peptides is a major hallmark of Alzheimer's disease (AD) and plays a crucial role in its pathogenesis. Particularly, the structured oligomeric species rich in β-sheet formations were implicated in neuronal organelle damage. Addressing this formidable challenge requires identifying candidates capable of inhibiting peptide aggregation or disaggregating preformed oligomers for effective antiaggregation-based AD therapy. Here, we present a dual-functional nanoinhibitor meticulously designed to target the aggregation driving force and amyloid fibril spatial structure. Leveraging the exceptional structural stability and facile tailoring capability of endohedral metallofullerene Gd@C82, we introduce desired hydrogen-binding sites and charged groups, which are abundant on its surface for specific designs. Impressively, these designs endow the resultant functionalized-Gd@C82 nanoparticles (f-Gd@C82 NPs) with high capability of redirecting peptide self-assembly toward disordered, off-pathway species, obstructing the early growth of protofibrils, and disaggregating the preformed well-ordered protofibrils or even mature Aβ fibrils. This results in considerable alleviation of Aβ peptide-induced neuronal cytotoxicity, rescuing neuronal death and synaptic loss in primary neuron models. Notably, these modifications significantly improved the dispersibility of f-Gd@C82 NPs, thus substantially enhancing its bioavailability. Moreover, f-Gd@C82 NPs demonstrate excellent cytocompatibility with various cell lines and possess the ability to penetrate the blood-brain barrier in mice. Large-scale molecular dynamics simulations illuminate the inhibition and disaggregation mechanisms. Our design successfully overcomes the limitations of other nanocandidates, which often overly rely on hydrophobic interactions or photothermal conversion properties, and offers a viable direction for developing anti-AD agents through the inhibition and even reversal of Aβ aggregation.
Collapse
Affiliation(s)
- Xiuhua Yin
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Hong Zhou
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Tiantian Cao
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
- Suzhou Institute of Trade and Commerce, Suzhou 215009, China
| | - Xiner Yang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Fei Meng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Xing Dai
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Yifan Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Sijie Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Wangsong Zhai
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Zaixing Yang
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Ning Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| |
Collapse
|
34
|
Polykretis P, D’Andrea C, Banchelli M, Napolitano L, Cascella R, de Angelis M, Matteini P. Exploring the Aβ 1-42 fibrillogenesis timeline by atomic force microscopy and surface enhanced Raman spectroscopy. Front Mol Biosci 2024; 11:1376411. [PMID: 38948077 PMCID: PMC11211275 DOI: 10.3389/fmolb.2024.1376411] [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: 01/25/2024] [Accepted: 05/22/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction: Alzheimer's disease (AD) is a progressive debilitating neurological disorder representing the most common neurodegenerative disease worldwide. Although the exact pathogenic mechanisms of AD remain unresolved, the presence of extracellular amyloid-β peptide 1-42 (Aβ1-42) plaques in the parenchymal and cortical brain is considered one of the hallmarks of the disease. Methods: In this work, we investigated the Aβ1-42 fibrillogenesis timeline up to 48 h of incubation, providing morphological and chemo-structural characterization of the main assemblies formed during the aggregation process of Aβ1-42, by atomic force microscopy (AFM) and surface enhanced Raman spectroscopy (SERS), respectively. Results: AFM topography evidenced the presence of characteristic protofibrils at early-stages of aggregation, which form peculiar macromolecular networks over time. SERS allowed to track the progressive variation in the secondary structure of the aggregation species involved in the fibrillogenesis and to determine when the β-sheet starts to prevail over the random coil conformation in the aggregation process. Discussion: Our research highlights the significance of investigating the early phases of fibrillogenesis to better understand the molecular pathophysiology of AD and identify potential therapeutic targets that may prevent or slow down the aggregation process.
Collapse
Affiliation(s)
- Panagis Polykretis
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| | - Cristiano D’Andrea
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| | - Martina Banchelli
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| | - Liliana Napolitano
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Marella de Angelis
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| | - Paolo Matteini
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| |
Collapse
|
35
|
Liu B, Li X, Liu Z, He B, Xu H, Cao J, Zeng F, Feng H, Ren Y, Li H, Wang T, Li J, Ye Y, Zhao L, Ran C, Li Y. Iterative Design of Near-Infrared Fluorescent Probes for Early Diagnosis of Alzheimer's Disease by Targeting Aβ Oligomers. J Med Chem 2024; 67:9104-9123. [PMID: 38829030 DOI: 10.1021/acs.jmedchem.4c00252] [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: 06/05/2024]
Abstract
Amyloid-β oligomers (AβOs), crucial toxic proteins in early Alzheimer's disease (AD), precede the formation of Aβ plaques and cognitive impairment. In this context, we present our iterative process for developing novel near-infrared fluorescent (NIRF) probes specifically targeting AβOs, aimed at early AD diagnosis. An initial screening identified compound 18 as being highly selective for AβOs. Subsequent analysis revealed that compound 20 improved serum stability while retaining affinity for AβOs. The most promising iteration, compound 37, demonstrated exceptional qualities: a high affinity for AβOs, emission in the near-infrared region, and good biocompatibility. Significantly, ex vivo double staining indicated that compound 37 detected AβOs in AD mouse brain and in vivo imaging experiments showed that compound 37 could differentiate between 4-month-old AD mice and age-matched wild-type mice. Therefore, compound 37 has emerged as a valuable NIRF probe for early detection of AD and a useful tool in exploring AD's pathological mechanisms.
Collapse
Affiliation(s)
- Bing Liu
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Xiaofang Li
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Zhengyang Liu
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Bing He
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Hanyue Xu
- Nanjing Foreign Language School, Nanjing 210008, Jiangsu, China
| | - Jianqin Cao
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Fantian Zeng
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Haiwei Feng
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Yanwei Ren
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Haoyu Li
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Tianyu Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Jia Li
- Pathology and PDX Efficacy Center, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Yuting Ye
- Pathology and PDX Efficacy Center, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Li Zhao
- School of Basic Medicine and Clinical Pharmacology, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Yuyan Li
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| |
Collapse
|
36
|
Mackness BC, Morgan BR, Deveau LM, Kathuria SV, Zitzewitz JA, Massi F. A hydrophobic core stabilizes the residual structure in the RRM2 intermediate state of the ALS-linked protein TDP-43. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598648. [PMID: 38915526 PMCID: PMC11195224 DOI: 10.1101/2024.06.12.598648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Folding intermediates mediate both protein folding and the misfolding and aggregation observed in human diseases, including amyotrophic lateral sclerosis (ALS), and are prime targets for therapeutic interventions. In this study, we identified the core nucleus of structure for a folding intermediate in the second RNA recognition motif (RRM2) of the ALS-linked RNA-binding protein, TDP-43, using a combination of experimental and computational approaches. Urea equilibrium unfolding studies revealed that the RRM2 intermediate state consists of collapsed residual secondary structure localized to the N-terminal half of RRM2, while the C-terminus is largely disordered. Steered molecular dynamics simulations and mutagenesis studies yielded key stabilizing hydrophobic contacts that, when mutated to alanine, severely disrupt the overall fold of RRM2. In combination, these findings suggest a role for this RRM intermediate in normal TDP-43 function as well as serving as a template for misfolding and aggregation through the low stability and non-native secondary structure.
Collapse
|
37
|
Balusu S, De Strooper B. The necroptosis cell death pathway drives neurodegeneration in Alzheimer's disease. Acta Neuropathol 2024; 147:96. [PMID: 38852117 PMCID: PMC11162975 DOI: 10.1007/s00401-024-02747-5] [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: 04/01/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Although apoptosis, pyroptosis, and ferroptosis have been implicated in AD, none fully explains the extensive neuronal loss observed in AD brains. Recent evidence shows that necroptosis is abundant in AD, that necroptosis is closely linked to the appearance of Tau pathology, and that necroptosis markers accumulate in granulovacuolar neurodegeneration vesicles (GVD). We review here the neuron-specific activation of the granulovacuolar mediated neuronal-necroptosis pathway, the potential AD-relevant triggers upstream of this pathway, and the interaction of the necrosome with the endo-lysosomal pathway, possibly providing links to Tau pathology. In addition, we underscore the therapeutic potential of inhibiting necroptosis in neurodegenerative diseases such as AD, as this presents a novel avenue for drug development targeting neuronal loss to preserve cognitive abilities. Such an approach seems particularly relevant when combined with amyloid-lowering drugs.
Collapse
Affiliation(s)
- Sriram Balusu
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, 3000, Leuven, Belgium.
- Leuven Brain Institute, KU Leuven, 3000, Leuven, Belgium.
| | - Bart De Strooper
- Laboratory for the Research of Neurodegenerative Diseases, VIB Center for Brain and Disease Research, 3000, Leuven, Belgium.
- Leuven Brain Institute, KU Leuven, 3000, Leuven, Belgium.
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK.
| |
Collapse
|
38
|
Nagashima K, Watanabe H, Akasaka T, Ono M. Development of Triphenylmethane Dyes for In Vivo Fluorescence Imaging of Aβ Oligomers. ACS Chem Neurosci 2024; 15:2233-2242. [PMID: 38753435 DOI: 10.1021/acschemneuro.4c00053] [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: 05/18/2024] Open
Abstract
Detection of amyloid β (Aβ) oligomers, regarded as the most toxic aggregated forms of Aβ, can contribute to the diagnosis and treatment of Alzheimer's disease (AD). Thus, the development of imaging probes for in vivo visualization of Aβ oligomers is crucial. However, the structural uncertainty regarding Aβ oligomers makes it difficult to design imaging probes with high sensitivity to Aβ oligomers against highly aggregated Aβ fibrils. In this study, we developed Aβ oligomer-selective fluorescent probes based on triphenylmethane dyes through screening of commercially available compounds followed by structure-activity relationship (SAR) studies on cyclic or acyclic 4-dialkylamino groups. We synthesized 11 triarylmethane-based Aβ oligomer probe (TAMAOP) derivatives. In vitro evaluation of fluorescence properties, TAMAOP-9, which had bulky 4-diisobutylamino groups introduced into three benzenes of a twisted triphenylmethane backbone, showed marked fluorescence enhancement in the presence of Aβ oligomers and demonstrated high selectivity for Aβ oligomers against Aβ fibrils. In docking studies using the Aβ trimer model, TAMAOP-9 bound to the hydrophobic surface and interacted with the side chain of Phe20. In vitro section staining revealed that TAMAOP-9 could visualize Aβ oligomers in the brains of AD model mice. An in vivo fluorescence imaging study using TAMAOP-9 showed significantly higher fluorescence signals from the brains of AD model mice than those of age-matched wild-type mice, confirmed by ex vivo section observation. These results suggest that TAMAOP-9 is a promising Aβ oligomer-targeting fluorescent probe applicable to in vivo imaging.
Collapse
Affiliation(s)
- Kotaro Nagashima
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takahiro Akasaka
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
39
|
Umar M, Rehman Y, Ambreen S, Mumtaz SM, Shaququzzaman M, Alam MM, Ali R. Innovative approaches to Alzheimer's therapy: Harnessing the power of heterocycles, oxidative stress management, and nanomaterial drug delivery system. Ageing Res Rev 2024; 97:102298. [PMID: 38604453 DOI: 10.1016/j.arr.2024.102298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/10/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Alzheimer's disease (AD) presents a complex pathology involving amyloidogenic proteolysis, neuroinflammation, mitochondrial dysfunction, and cholinergic deficits. Oxidative stress exacerbates AD progression through pathways like macromolecular peroxidation, mitochondrial dysfunction, and metal ion redox potential alteration linked to amyloid-beta (Aβ). Despite limited approved medications, heterocyclic compounds have emerged as promising candidates in AD drug discovery. This review highlights recent advancements in synthetic heterocyclic compounds targeting oxidative stress, mitochondrial dysfunction, and neuroinflammation in AD. Additionally, it explores the potential of nanomaterial-based drug delivery systems to overcome challenges in AD treatment. Nanoparticles with heterocyclic scaffolds, like polysorbate 80-coated PLGA and Resveratrol-loaded nano-selenium, show improved brain transport and efficacy. Micellar CAPE and Melatonin-loaded nano-capsules exhibit enhanced antioxidant properties, while a tetra hydroacridine derivative (CHDA) combined with nano-radiogold particles demonstrates promising acetylcholinesterase inhibition without toxicity. This comprehensive review underscores the potential of nanotechnology-driven drug delivery for optimizing the therapeutic outcomes of novel synthetic heterocyclic compounds in AD management. Furthermore, the inclusion of various promising heterocyclic compounds with detailed ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) data provides valuable insights for planning the development of novel drug delivery treatments for AD.
Collapse
Affiliation(s)
- Mohammad Umar
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi 110017, India
| | - Yasir Rehman
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Subiya Ambreen
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi 110017, India
| | - Sayed Md Mumtaz
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Mohd Shaququzzaman
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Mohammad Mumtaz Alam
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi 110017, India.
| |
Collapse
|
40
|
Qiu Y, Huang T, Cai YD. Review of predicting protein stability changes upon variations. Proteomics 2024; 24:e2300371. [PMID: 38643379 DOI: 10.1002/pmic.202300371] [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: 01/30/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/22/2024]
Abstract
Forecasting alterations in protein stability caused by variations holds immense importance. Improving the thermal stability of proteins is important for biomedical and industrial applications. This review discusses the latest methods for predicting the effects of mutations on protein stability, databases containing protein mutations and thermodynamic parameters, and experimental techniques for efficiently assessing protein stability in high-throughput settings. Various publicly available databases for protein stability prediction are introduced. Furthermore, state-of-the-art computational approaches for anticipating protein stability changes due to variants are reviewed. Each method's types of features, base algorithm, and prediction results are also detailed. Additionally, some experimental approaches for verifying the prediction results of computational methods are introduced. Finally, the review summarizes the progress and challenges of protein stability prediction and discusses potential models for future research directions.
Collapse
Affiliation(s)
- Yiling Qiu
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Mathematics and Statistics, Guangdong University of Technology, Guangzhou, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
41
|
Zhang H, Cai W, Dong L, Yang Q, Li Q, Ran Q, Liu L, Wang Y, Li Y, Weng X, Zhu X, Chen Y. Jiaohong pills attenuate neuroinflammation and amyloid-β protein-induced cognitive deficits by modulating the mitogen-activated protein kinase/nuclear factor kappa-B pathway. Animal Model Exp Med 2024; 7:222-233. [PMID: 38177948 PMCID: PMC11228096 DOI: 10.1002/ame2.12369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 11/15/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Jiaohong pills (JHP) consist of Pericarpium Zanthoxyli (PZ) and Radix Rehmanniae, two herbs that have been extensively investigated over many years due to their potential protective effects against cognitive decline and memory impairment. However, the precise mechanisms underlying the beneficial effects remain elusive. Here, research studies were conducted to investigate and validate the therapeutic effects of JHP on Alzheimer's disease. METHODS BV-2 cell inflammation was induced by lipopolysaccharide. AD mice were administered amyloid-β (Aβ). Behavioral experiments were used to evaluate learning and memory ability. The levels of nitric oxide (NO), tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-10 (IL-10) were detected using enzyme-linked immunosorbent assay (ELISA). The protein expressions of inducible nitric oxide synthase (iNOS) and the phosphorylation level of mitogen-activated protein kinase (MAPK) and nuclear factor kappa-B (NF-κB) were detected using Western blot. Nissl staining was used to detect neuronal degeneration. RESULTS The results demonstrated that an alcoholic extract of PZ significantly decreased the levels of NO, IL-1β, TNF-α, and iNOS; increased the expression level of IL-10; and significantly decreased the phosphorylation levels of MAPK and NF-κB. These inhibitory effects were further confirmed in the AD mouse model. Meanwhile, JHP improved learning and memory function in AD mice, reduced neuronal damage, and enriched the Nissl bodies in the hippocampus. Moreover, IL-1β and TNF-α in the cortex were significantly downregulated after JHP administration, whereas IL-10 showed increased expression. CONCLUSIONS It was found that JHP reduced neuroinflammatory response in AD mice by targeting the MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Hong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weiyan Cai
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lijinchuan Dong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qing Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingsen Ran
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yajie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaogang Weng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Xia Z, Prescott EE, Urbanek A, Wareing HE, King MC, Olerinyova A, Dakin H, Leah T, Barnes KA, Matuszyk MM, Dimou E, Hidari E, Zhang YP, Lam JYL, Danial JSH, Strickland MR, Jiang H, Thornton P, Crowther DC, Ohtonen S, Gómez-Budia M, Bell SM, Ferraiuolo L, Mortiboys H, Higginbottom A, Wharton SB, Holtzman DM, Malm T, Ranasinghe RT, Klenerman D, De S. Co-aggregation with Apolipoprotein E modulates the function of Amyloid-β in Alzheimer's disease. Nat Commun 2024; 15:4695. [PMID: 38824138 PMCID: PMC11144216 DOI: 10.1038/s41467-024-49028-z] [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: 02/28/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024] Open
Abstract
Which isoforms of apolipoprotein E (apoE) we inherit determine our risk of developing late-onset Alzheimer's Disease (AD), but the mechanism underlying this link is poorly understood. In particular, the relevance of direct interactions between apoE and amyloid-β (Aβ) remains controversial. Here, single-molecule imaging shows that all isoforms of apoE associate with Aβ in the early stages of aggregation and then fall away as fibrillation happens. ApoE-Aβ co-aggregates account for ~50% of the mass of diffusible Aβ aggregates detected in the frontal cortices of homozygotes with the higher-risk APOE4 gene. We show how dynamic interactions between apoE and Aβ tune disease-related functions of Aβ aggregates throughout the course of aggregation. Our results connect inherited APOE genotype with the risk of developing AD by demonstrating how, in an isoform- and lipidation-specific way, apoE modulates the aggregation, clearance and toxicity of Aβ. Selectively removing non-lipidated apoE4-Aβ co-aggregates enhances clearance of toxic Aβ by glial cells, and reduces secretion of inflammatory markers and membrane damage, demonstrating a clear path to AD therapeutics.
Collapse
Affiliation(s)
- Zengjie Xia
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Emily E Prescott
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Agnieszka Urbanek
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Hollie E Wareing
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Marianne C King
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Anna Olerinyova
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Helen Dakin
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
- Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Tom Leah
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Katy A Barnes
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Martyna M Matuszyk
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Eleni Dimou
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Eric Hidari
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Yu P Zhang
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - Jeff Y L Lam
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
| | - John S H Danial
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK
- SUPA School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, KY16 9SS, UK
| | - Michael R Strickland
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hong Jiang
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter Thornton
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Sohvi Ohtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mireia Gómez-Budia
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Simon M Bell
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
- Healthy Lifespan Institute (HELSI), University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Rohan T Ranasinghe
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK.
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
- UK Dementia Research Institute at University of Cambridge, Cambridge, UK.
| | - Suman De
- Sheffield Institute for Translational Neuroscience, Division of Neurosciences, University of Sheffield, Sheffield, S10 2HQ, UK.
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK.
- Healthy Lifespan Institute (HELSI), University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
43
|
Kaku T, Ikebukuro K, Tsukakoshi K. Structure of cytotoxic amyloid oligomers generated during disaggregation. J Biochem 2024; 175:575-585. [PMID: 38430131 PMCID: PMC11155694 DOI: 10.1093/jb/mvae023] [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: 08/30/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
Amyloidosis is characterized by the abnormal accumulation of amyloid proteins. The causative proteins aggregate from monomers to oligomers and fibrils, among which some intermediate oligomers are considered as major toxins. Cytotoxic oligomers are generated not only by aggregation but also via fibril disaggregation. However, little is known about the structural characteristics and generation conditions of cytotoxic oligomers produced during disaggregation. Herein, we summarized the structural commonalities of cytotoxic oligomers formed under various disaggregation conditions, including the addition of heat shock proteins or small compounds. In vitro experimental data demonstrated the presence of high-molecular-weight oligomers (protofibrils or protofilaments) that exhibited a fibrous morphology and β-sheet structure. Molecular dynamics simulations indicated that the distorted β-sheet structure contributed to their metastability. The tendency of these cytotoxic oligomers to appear under mild disaggregation conditions, implied formation during the early stages of disaggregation. This review will aid researchers in exploring the characteristics of highly cytotoxic oligomers and developing drugs that target amyloid aggregates.
Collapse
Affiliation(s)
- Toshisuke Kaku
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kazunori Ikebukuro
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kaori Tsukakoshi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| |
Collapse
|
44
|
Zhang L, Cao K, Xie J, Liang X, Gong H, Luo Q, Luo H. Aβ 42 and ROS dual-targeted multifunctional nanocomposite for combination therapy of Alzheimer's disease. J Nanobiotechnology 2024; 22:278. [PMID: 38783363 PMCID: PMC11112798 DOI: 10.1186/s12951-024-02543-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Amyloid-β (Aβ) readily misfolds into neurotoxic aggregates, generating high levels of reactive oxygen species (ROS), leading to progressive oxidative damage and ultimately cell death. Therefore, simultaneous inhibition of Aβ aggregation and scavenging of ROS may be a promising therapeutic strategy to alleviate Alzheimer's disease pathology. Based on the previously developed antibody 1F12 that targets all forms of Aβ42, we developed an Aβ42 and ROS dual-targeting nanocomposite using biodegradable mesoporous silica nanoparticles as carriers to load ultra-small cerium oxide nanocrystals (bMSNs@Ce-1F12). By modifying the brain-targeted rabies virus glycoprotein 29 (RVG29-bMSNs@Ce-1F12), this intelligent nanocomposite can efficiently target brain Aβ-rich regions. Combined with peripheral and central nervous system treatments, RVG29-bMSNs@Ce-1F12 can significantly alleviate AD symptoms by inhibiting Aβ42 misfolding, accelerating Aβ42 clearance, and scavenging ROS. Furthermore, this synergistic effect of ROS scavenging and Aβ clearance exhibited by this Aβ42 and ROS dual-targeted strategy also reduced the burden of hyperphosphorylated tau, alleviated glial cell activation, and ultimately improved cognitive function in APP/PS1 mice. Our findings indicate that RVG29-bMSNs@Ce-1F12 is a promising nanodrug that can facilitate multi-target treatment of AD.
Collapse
Affiliation(s)
- Liding Zhang
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, 570228, China
| | - Kai Cao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jun Xie
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaohan Liang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, 215123, China
| | - Qingming Luo
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, 570228, China.
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, 215123, China.
| | - Haiming Luo
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, 570228, China.
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, 215123, China.
| |
Collapse
|
45
|
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
|
46
|
Meng X, Song Q, Liu Z, Liu X, Wang Y, Liu J. Neurotoxic β-amyloid oligomers cause mitochondrial dysfunction-the trigger for PANoptosis in neurons. Front Aging Neurosci 2024; 16:1400544. [PMID: 38808033 PMCID: PMC11130508 DOI: 10.3389/fnagi.2024.1400544] [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: 03/13/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
As the global population ages, the incidence of elderly patients with dementia, represented by Alzheimer's disease (AD), will continue to increase. Previous studies have suggested that β-amyloid protein (Aβ) deposition is a key factor leading to AD. However, the clinical efficacy of treating AD with anti-Aβ protein antibodies is not satisfactory, suggesting that Aβ amyloidosis may be a pathological change rather than a key factor leading to AD. Identification of the causes of AD and development of corresponding prevention and treatment strategies is an important goal of current research. Following the discovery of soluble oligomeric forms of Aβ (AβO) in 1998, scientists began to focus on the neurotoxicity of AβOs. As an endogenous neurotoxin, the active growth of AβOs can lead to neuronal death, which is believed to occur before plaque formation, suggesting that AβOs are the key factors leading to AD. PANoptosis, a newly proposed concept of cell death that includes known modes of pyroptosis, apoptosis, and necroptosis, is a form of cell death regulated by the PANoptosome complex. Neuronal survival depends on proper mitochondrial function. Under conditions of AβO interference, mitochondrial dysfunction occurs, releasing lethal contents as potential upstream effectors of the PANoptosome. Considering the critical role of neurons in cognitive function and the development of AD as well as the regulatory role of mitochondrial function in neuronal survival, investigation of the potential mechanisms leading to neuronal PANoptosis is crucial. This review describes the disruption of neuronal mitochondrial function by AβOs and elucidates how AβOs may activate neuronal PANoptosis by causing mitochondrial dysfunction during the development of AD, providing guidance for the development of targeted neuronal treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Jinyu Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
47
|
Al Khashali H, Ray R, Darweesh B, Wozniak C, Haddad B, Goel S, Seidu I, Khalil J, Lopo B, Murshed N, Guthrie J, Heyl D, Evans HG. Amyloid Beta Leads to Decreased Acetylcholine Levels and Non-Small Cell Lung Cancer Cell Survival via a Mechanism That Involves p38 Mitogen-Activated Protein Kinase and Protein Kinase C in a p53-Dependent and -Independent Manner. Int J Mol Sci 2024; 25:5033. [PMID: 38732252 PMCID: PMC11084752 DOI: 10.3390/ijms25095033] [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: 04/11/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024] Open
Abstract
Several studies have shown an inverse correlation between the likelihood of developing a neurodegenerative disorder and cancer. We previously reported that the levels of amyloid beta (Aβ), at the center of Alzheimer's disease pathophysiology, are regulated by acetylcholinesterase (AChE) in non-small cell lung cancer (NSCLC). Here, we examined the effect of Aβ or its fragments on the levels of ACh in A549 (p53 wild-type) and H1299 (p53-null) NSCLC cell media. ACh levels were reduced by cell treatment with Aβ 1-42, Aβ 1-40, Aβ 1-28, and Aβ 25-35. AChE and p53 activities increased upon A549 cell treatment with Aβ, while knockdown of p53 in A549 cells increased ACh levels, decreased AChE activity, and diminished the Aβ effects. Aβ increased the ratio of phospho/total p38 MAPK and decreased the activity of PKC. Inhibiting p38 MAPK reduced the activity of p53 in A549 cells and increased ACh levels in the media of both cell lines, while opposite effects were found upon inhibiting PKC. ACh decreased the activity of p53 in A549 cells, decreased p38 MAPK activity, increased PKC activity, and diminished the effect of Aβ on those activities. Moreover, the negative effect of Aβ on cell viability was diminished by cell co-treatment with ACh.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Hedeel Guy Evans
- Chemistry Department, Eastern Michigan University, Ypsilanti, MI 48197, USA; (H.A.K.); (R.R.); (B.D.); (C.W.); (B.H.); (S.G.); (I.S.); (J.K.); (B.L.); (N.M.); (J.G.); (D.H.)
| |
Collapse
|
48
|
Basir HS, Mirazi N, Komaki A, Hosseini A. Cacao consumption improves passive avoidance memory impairment in a rat model of Alzheimer's disease: the role of hippocampal synaptic plasticity and oxidative stress. Front Pharmacol 2024; 15:1379264. [PMID: 38756381 PMCID: PMC11096498 DOI: 10.3389/fphar.2024.1379264] [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: 01/30/2024] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction: Alzheimer's disease (AD) causes progressive loss of cognitive function and synaptic plasticity, which is the most common form of dementia. The present study was designed to scrutinize the effects of cacao on passive avoidance memory function and to identify the roles of hippocampal synaptic plasticity and oxidative stress in an AD rat model induced by unilateral intracerebroventricular (UICV) injection of amyloid-beta (Aβ). Methods: Oral administration of cacao (500 mg/kg/ day) was given for 2 consecutive months. A memory retention test was conducted 24 h after passive avoidance training was completed. Subsequently, the amplitude of population spike (PS) and slope of field excitatory postsynaptic potentials (fEPSPs) were assessed at hippocampal long-term potentiation (LTP) in perforant pathway-dentate gyrus (PP-DG) synapses. Moreover, total thiol group (TTG) and malondialdehyde (MDA) concentrations were evaluated in the plasma. Furthermore, compact Aβ plaques were detected in the hippocampal DG by performing Congo red staining. Results: As a result of AD induction, passive avoidance memory was impaired; also, reduced fEPSP slopes, PS amplitudes, and content of TTG, and increase in MDA levels in the rats were observed. In contrast, cacao treatment ameliorated passive avoidance memory impairment, improved hippocampal LTP impairment, modulated oxidative-antioxidative status, and delayed Aβ plaques production in AD rats. Disscussion: Conclusively, cacao alleviates Aβ-induced cognitive deficit, probably by the amelioration of hippocampal LTP impairment, modulation of oxidative-antioxidative status, and inhibition of Aβ plaque accumulation.
Collapse
Affiliation(s)
- Hamid Shokati Basir
- Department of Biology, Faculty of Basic Science, Bu-Ali Sina University, Hamedan, Iran
| | - Naser Mirazi
- Department of Biology, Faculty of Basic Science, Bu-Ali Sina University, Hamedan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolkarim Hosseini
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
49
|
Tapella L, Dematteis G, La Vitola P, Leva S, Tonelli E, Raddi M, Delconti M, Dacomo L, La Macchia A, Murari E, Talmon M, Malecka J, Chrostek G, Grilli M, Colombo L, Salmona M, Forloni G, Genazzani AA, Balducci C, Lim D. Genetic deletion of astrocytic calcineurin B1 prevents cognitive impairment and neuropathology development in acute and chronic mouse models of Alzheimer's disease. Glia 2024; 72:899-915. [PMID: 38288580 DOI: 10.1002/glia.24509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 03/20/2024]
Abstract
Alzheimer's disease (AD) represents an urgent yet unmet challenge for modern society, calling for exploration of innovative targets and therapeutic approaches. Astrocytes, main homeostatic cells in the CNS, represent promising cell-target. Our aim was to investigate if deletion of the regulatory CaNB1 subunit of calcineurin in astrocytes could mitigate AD-related memory deficits, neuropathology, and neuroinflammation. We have generated two, acute and chronic, AD mouse models with astrocytic CaNB1 ablation (ACN-KO). In the former, we evaluated the ability of β-amyloid oligomers (AβOs) to impair memory and activate glial cells once injected in the cerebral ventricle of conditional ACN-KO mice. Next, we generated a tamoxifen-inducible astrocyte-specific CaNB1 knock-out in 3xTg-AD mice (indACNKO-AD). CaNB1 was deleted, by tamoxifen injection, in 11.7-month-old 3xTg-AD mice for 4.4 months. Spatial memory was evaluated using the Barnes maze; β-amyloid plaques burden, neurofibrillary tangle deposition, reactive gliosis, and neuroinflammation were also assessed. The acute model showed that ICV injected AβOs in 2-month-old wild type mice impaired recognition memory and fostered a pro-inflammatory microglia phenotype, whereas in ACN-KO mice, AβOs were inactive. In indACNKO-AD mice, 4.4 months after CaNB1 depletion, we found preservation of spatial memory and cognitive flexibility, abolishment of amyloidosis, and reduction of neurofibrillary tangles, gliosis, and neuroinflammation. Our results suggest that ACN is crucial for the development of cognitive impairment, AD neuropathology, and neuroinflammation. Astrocyte-specific CaNB1 deletion is beneficial for both the abolishment of AβO-mediated detrimental effects and treatment of ongoing AD-related pathology, hence representing an intriguing target for AD therapy.
Collapse
Affiliation(s)
- Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Pietro La Vitola
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Susanna Leva
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Elisa Tonelli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Marco Raddi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Marta Delconti
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Letizia Dacomo
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Alberto La Macchia
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Elisa Murari
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Maria Talmon
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Justyna Malecka
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Gabriela Chrostek
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Laura Colombo
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Mario Salmona
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Claudia Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
50
|
Mancuso R, Fattorelli N, Martinez-Muriana A, Davis E, Wolfs L, Van Den Daele J, Geric I, Premereur J, Polanco P, Bijnens B, Preman P, Serneels L, Poovathingal S, Balusu S, Verfaillie C, Fiers M, De Strooper B. Xenografted human microglia display diverse transcriptomic states in response to Alzheimer's disease-related amyloid-β pathology. Nat Neurosci 2024; 27:886-900. [PMID: 38539015 PMCID: PMC11089003 DOI: 10.1038/s41593-024-01600-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/14/2024] [Indexed: 05/14/2024]
Abstract
Microglia are central players in Alzheimer's disease pathology but analyzing microglial states in human brain samples is challenging due to genetic diversity, postmortem delay and admixture of pathologies. To circumvent these issues, here we generated 138,577 single-cell expression profiles of human stem cell-derived microglia xenotransplanted in the brain of the AppNL-G-F model of amyloid pathology and wild-type controls. Xenografted human microglia adopt a disease-associated profile similar to that seen in mouse microglia, but display a more pronounced human leukocyte antigen or HLA state, likely related to antigen presentation in response to amyloid plaques. The human microglial response also involves a pro-inflammatory cytokine/chemokine cytokine response microglia or CRM response to oligomeric Aβ oligomers. Genetic deletion of TREM2 or APOE as well as APOE polymorphisms and TREM2R47H expression in the transplanted microglia modulate these responses differentially. The expression of other Alzheimer's disease risk genes is differentially regulated across the distinct cell states elicited in response to amyloid pathology. Thus, we have identified multiple transcriptomic cell states adopted by human microglia in a multipronged response to Alzheimer's disease-related pathology, which should be taken into account in translational studies.
Collapse
Affiliation(s)
- Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium.
| | - Nicola Fattorelli
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Anna Martinez-Muriana
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Emma Davis
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Leen Wolfs
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Johanna Van Den Daele
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Leuven, Belgium
| | - Ivana Geric
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Jessie Premereur
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Paula Polanco
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Baukje Bijnens
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Pranav Preman
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Lutgarde Serneels
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Suresh Poovathingal
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
| | - Sriram Balusu
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Catherine Verfaillie
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Leuven, Belgium
| | - Mark Fiers
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Bart De Strooper
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium.
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium.
- UK Dementia Research Institute at UCL, University College London, London, UK.
| |
Collapse
|