101
|
Wang M, Jin B, Jo J. Acute Restraint Stress Induces Long-Lasting Synaptic Enhancement by Inhibiting AMPK Activation in AD Model Mice. CNS Neurosci Ther 2025; 31:e70335. [PMID: 40102200 PMCID: PMC11919636 DOI: 10.1111/cns.70335] [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: 07/13/2024] [Revised: 11/18/2024] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by a gradual synaptic loss. The progression of AD severely affects late-phase long-term potentiation (L-LTP), which is essential for long-term memory consolidation. AIM We have previously demonstrated the beneficial effects of acute restraint stress (ARS) on hippocampal LTP in AD mouse models. This study aimed to verify the effects and potential mechanisms of ARS on the maintenance of hippocampal L-LTP in two AD mouse models. MATERIALS AND METHODS 5xFAD and Tg2576 mice underwent a 30-min body immobilization protocol to induce ARS, followed by electrophysiological recordings of L-LTP (> 3 h) in the CA1 region of thehippocampus. RESULTS The ARS-exposed group exhibited significantly enhanced L-LTP compared to the control group. Maintenance of L-LTP requires new protein synthesis and signaling via the mammalian target of rapamycin (mTOR) pathway. Our findings revealed that ARS increased hippocampal adenosine triphosphate (ATP) production and reduced AMPK activity. Inactivation of AMPK and subsequent activation of the mTOR pathway were strongly associated with the ARS-facilitated enhancement of L-LTP. Furthermore, our experiments using the mTOR inhibitor rapamycin demonstrated that it effectively prevented the enhancement of L-LTP following ARS, underscoring the pivotal role of mTOR in this process. CONCLUSION ARS may significantly modify AMPK activation and mTOR regulation in L-LTP, potentially triggering the mechanisms of long-term memory consolidation in AD mouse model mice. Identifying these underlying mechanisms could help promote the development of novel pharmaceutical agents for the treatment of AD.
Collapse
Affiliation(s)
- Ming Wang
- School of Public Health, Health Science Center, Ningbo University, Ningbo, China
| | - Baoyuan Jin
- The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Jihoon Jo
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
102
|
Zhang Z, Zhang Y, Peng H, Yu Q, Kang X, Liu Y, Zheng Y, Cheng F, Wang X, Li F. Decoding TGR5: A comprehensive review of its impact on cerebral diseases. Pharmacol Res 2025; 213:107671. [PMID: 39988005 DOI: 10.1016/j.phrs.2025.107671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Currently, unraveling the enigmatic realm of drug targets for cerebral disorders poses a formidable challenge. Takeda G protein-coupled receptor 5 (TGR5), also known as G protein-coupled bile acid receptor 1, is a specific bile acid receptor. Widely distributed across various tissues, TGR5 orchestrates a myriad of biological functions encompassing inflammation, energy metabolism, fatty acid metabolism, immune responses, cellular proliferation, apoptosis, and beyond. Alongside its well-documented implications in liver diseases, obesity, type 2 diabetes, tumors, and cardiovascular diseases, a growing body of evidence accentuates the pivotal role of TGR5 in cerebral diseases. Thus, this comprehensive review aimed to scrutinize the current insights into the pathological mechanisms involving TGR5 in cerebral diseases, while contemplating its potential as a promising therapeutic target for cerebral diseases.
Collapse
Affiliation(s)
- Zehan Zhang
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Yifei Zhang
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Hongye Peng
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Qingqian Yu
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Xiangdong Kang
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Ying Liu
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Yuxiao Zheng
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Fafeng Cheng
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Xueqian Wang
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| | - Feng Li
- Beijing University of Chinese Medicine, The northeast corner of the intersection of Sunshine South Street and Baiyang East Road, Beijing 102488, China.
| |
Collapse
|
103
|
Hales CM. Alzheimer's Disease Diagnosis and Management in the Age of Amyloid Monoclonal Antibodies. Med Clin North Am 2025; 109:463-483. [PMID: 39893023 DOI: 10.1016/j.mcna.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, and for providers the term AD is often avoided, favoring generic terms like memory loss or dementia. This is partly not only by limitations in using diagnostics and busy clinics but also by a sense that an AD diagnosis will not lead to a meaningful change in management. However, a turning point has occurred with advancements in diagnostics and disease-modifying therapies. Additionally, AD prevention therapies are not too far into the future. This review will cover AD clinical presentation and symptomatic management with focus on AD diagnostics and disease-modifying therapies.
Collapse
Affiliation(s)
- Chadwick M Hales
- Department of Neurology, Center for Neurodegenerative Disease, Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, 6 Executive Park Drive, Atlanta, GA 30329, USA.
| |
Collapse
|
104
|
Djebari S, Jiménez-Herrera R, Iborra-Lázaro G, Jiménez-Díaz L, Navarro-López JD. Social and contextual memory impairments induced by Amyloid-β oligomers are rescued by Sigma-1 receptor activation. Biomed Pharmacother 2025; 184:117914. [PMID: 39999645 DOI: 10.1016/j.biopha.2025.117914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/06/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Sigma-1 receptors (S1Rs) are widely expressed throughout the central nervous system and modulate neuron intracellular calcium levels, leading to changes in neurotransmitter release and neuronal activity. They also interact with various proteins and signaling pathways, playing a key role in regulating synaptic plasticity in brain areas such as the hippocampus, thereby influencing learning and memory processes. This opens a research avenue to explore S1R modulation as a potential therapeutic target in diseases involving hippocampal synaptic alterations and compromised cognitive processes, such as Alzheimer's disease (AD). Here, we hypothesize that pharmacological activation of S1R could counteract synaptic plasticity deficits and hippocampal-dependent cognitive alterations in an early-stage amyloidosis model of Alzheimer's disease, induced by intracerebroventricular (icv) administration of Aβ1-42 oligomers (oAβ1-42). For that purpose, we investigate ex vivo CA3-CA1 synaptic plasticity, while in vivo, we performed open field habituation and social recognition tasks to assess contextual and social memory, respectively. Our data show that pharmacological activation of S1Rs with the selective agonist PRE-084 counteract oAβ1-42 deleterious effects on CA3-CA1 long-term synaptic plasticity (LTP), and hippocampal-dependent contextual and social memory, without alterations of spontaneous behaviors. Together, these results provide further evidence for the role of S1Rs in ameliorating hippocampal synaptic and contextual memory dysfunctions and introduce novel insight into their involvement in early amyloid-induced social memory deficits, highlighting their potential for developing comprehensive treatments for early AD. Also, the absence of adverse behavioral outcomes associated with PRE-084 treatment suggests a favorable safety profile in preclinical models, supporting its potential as a therapeutic option.
Collapse
Affiliation(s)
- Souhail Djebari
- Neurophysiology & Behavior Lab, Institute of Biomedicine (IB-UCLM) and Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Raquel Jiménez-Herrera
- Neurophysiology & Behavior Lab, Institute of Biomedicine (IB-UCLM) and Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Guillermo Iborra-Lázaro
- Neurophysiology & Behavior Lab, Institute of Biomedicine (IB-UCLM) and Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Lydia Jiménez-Díaz
- Neurophysiology & Behavior Lab, Institute of Biomedicine (IB-UCLM) and Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain.
| | - Juan D Navarro-López
- Neurophysiology & Behavior Lab, Institute of Biomedicine (IB-UCLM) and Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), Faculty of Medicine of Ciudad Real, University of Castilla-La Mancha, Ciudad Real, Spain.
| |
Collapse
|
105
|
Junyi L, Yueyang W, Bin L, Xiaohong D, Wenhui C, Ning Z, Hong Z. Gut Microbiota Mediates Neuroinflammation in Alzheimer's Disease: Unraveling Key Factors and Mechanistic Insights. Mol Neurobiol 2025; 62:3746-3763. [PMID: 39317889 DOI: 10.1007/s12035-024-04513-w] [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: 03/24/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
The gut microbiota, the complex community of microorganisms that inhabit the gastrointestinal tract, has emerged as a key player in the pathogenesis of neurodegenerative disorders, including Alzheimer's disease (AD). AD is characterized by progressive cognitive decline and neuronal loss, associated with the accumulation of amyloid-β plaques, neurofibrillary tangles, and neuroinflammation in the brain. Increasing evidence suggests that alterations in the composition and function of the gut microbiota, known as dysbiosis, may contribute to the development and progression of AD by modulating neuroinflammation, a chronic and maladaptive immune response in the central nervous system. This review aims to comprehensively analyze the current role of the gut microbiota in regulating neuroinflammation and glial cell function in AD. Its objective is to deepen our understanding of the pathogenesis of AD and to discuss the potential advantages and challenges of using gut microbiota modulation as a novel approach for the diagnosis, treatment, and prevention of AD.
Collapse
Affiliation(s)
- Liang Junyi
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Wang Yueyang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Liu Bin
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China.
| | - Dong Xiaohong
- Jiamusi College, Heilongjiang University of Traditional Chinese Medicine, Jiamusi, Heilongjiang Province, China
| | - Cai Wenhui
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Zhang Ning
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Zhang Hong
- Heilongjiang Jiamusi Central Hospital, Jiamusi, Heilongjiang Province, China
| |
Collapse
|
106
|
Ma X, Prokopenko D, Wang N, Aikawa T, Choi Y, Zhang C, Lei D, Ren Y, Kawatani K, Starling SC, Perkerson RB, Roy B, Quintero AC, Parsons TM, Pan Y, Li Z, Wang M, Bao H, Han X, Bu G, Tanzi RE, Kanekiyo T. Alzheimer's disease risk ABCA7 p.A696S variant disturbs the microglial response to amyloid pathology in mice. Neurobiol Dis 2025; 206:106813. [PMID: 39880319 PMCID: PMC11884743 DOI: 10.1016/j.nbd.2025.106813] [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: 08/31/2024] [Revised: 12/16/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
The adenosine triphosphate-binding cassette transporter A7 (ABCA7) gene is ranked as one of the top susceptibility loci for Alzheimer's disease (AD). While ABCA7 mediates lipid transport across cellular membranes, ABCA7 loss of function has been shown to exacerbate amyloid-β (Aβ) pathology and compromise microglial function. Our family-based study uncovered an extremely rare ABCA7 p.A696S variant that was substantially segregated with the development of AD in 3 African American families. Using the knockin mouse model, we investigated the effects of ABCA7-A696S substitution on amyloid pathology and brain immune response in 5xFAD transgenic mice. Importantly, our study demonstrated that ABCA7-A696S substitution reduces amyloid plaque-associated microgliosis and increases dystrophic neurites around amyloid deposits compared to control mice. We also found increased X-34-positive amyloid plaque burden in 5xFAD mice with ABCA7-A696S substitution, while there was no evident difference in insoluble Aβ levels between mouse groups. Thus, ABCA7-A696S substitution may disrupt amyloid compaction resulting in aggravated neuritic dystrophy due to insufficient microglia barrier function. In addition, we observed that ABCA7-A696S substitution disturbs the induction of proinflammatory cytokines interleukin 1β and interferon γ in the brains of 5xFAD mice, although some disease-associated microglia gene expression, including Trem2 and Tyrobp, are upregulated. Lipidomics also detected higher total lysophosphatidylethanolamine levels in the brains of 5xFAD mice with ABCA7-A696S substitution than controls. These results suggest that ABCA7-A696S substitution might compromise the adequate innate immune response to amyloid pathology in AD by modulating brain lipid metabolism, providing novel insight into the pathogenic mechanisms mediated by ABCA7. ONE SENTENCE SUMMARY: A rare Alzheimer's disease risk ABCA7 p.A696S variant compromises microglial response to amyloid pathology.
Collapse
Affiliation(s)
- Xiaoye Ma
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dmitry Prokopenko
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Ni Wang
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tomonori Aikawa
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Younjung Choi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Dan Lei
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Yingxue Ren
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Keiji Kawatani
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Skylar C Starling
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ralph B Perkerson
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Bhaskar Roy
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Astrid C Quintero
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tammee M Parsons
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yining Pan
- Department of Health Outcomes & Biomedical Informatics, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Zonghua Li
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hanmei Bao
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Xianlin Han
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Guojun Bu
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA.
| | - Takahisa Kanekiyo
- Departments of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
107
|
Kedia S, Simons M. Oligodendrocytes in Alzheimer's disease pathophysiology. Nat Neurosci 2025; 28:446-456. [PMID: 39881195 DOI: 10.1038/s41593-025-01873-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
Our understanding of Alzheimer's disease (AD) has transformed from a purely neuronal perspective to one that acknowledges the involvement of glial cells. Despite remarkable progress in unraveling the biology of microglia, astrocytes and vascular elements, the exploration of oligodendrocytes in AD is still in its early stages. Contrary to the traditional notion of oligodendrocytes as passive bystanders in AD pathology, emerging evidence indicates their active participation in and reaction to amyloid and tau pathology. Oligodendrocytes undergo a functional transition to a disease-associated state, engaging in immune modulation, stress responses and cellular survival. Far from being inert players, they appear to serve a dual role in AD pathogenesis, potentially offering defense mechanisms against pathology while also contributing to disease progression. This Review explores recent advancements in understanding the roles of oligodendrocytes and their myelin sheaths in the context of AD, shedding light on their complex interactions within the disease pathology.
Collapse
Affiliation(s)
- Shreeya Kedia
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
108
|
Dileep KV, Sakai N, Ihara K, Nakata A, Ito A, Sivaraman DM, Yip CW, Shin JW, Yoshida M, Shirouzu M, Zhang KYJ. Identification of benzimidazole-6-carboxamide based inhibitors of secretory glutaminyl cyclase for the treatment of Alzheimer's disease. Int J Biol Macromol 2025; 293:139320. [PMID: 39740711 DOI: 10.1016/j.ijbiomac.2024.139320] [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: 08/09/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/02/2025]
Abstract
The formation of the pyroglutamate variant of amyloid beta (pGlu-Aβ), which is extremely hydrophobic, rapidly aggregating, and highly neurotoxic, is mediated by the action of secretory glutaminyl cyclase (sQC). The pGlu-Aβ often acts as a seed for the aggregation of the full length Aβ and contributes to the overall load of Aβ plaques in Alzheimer's disease (AD). Therefore, inhibiting sQC is a potential approach to limit the formation of pGlu-Aβ and to modify the progression of AD. This study presents two novel molecules containing benzimidazole-6-carboxamide, namely LSB-09 and LSB-24, as promising sQC inhibitors. These inhibitors demonstrated moderate toxicity in human neuroblastoma cell lines and possessed IC50 values in the micromolar range (40 and 4 μM for LSB-09 and LSB-24, respectively). Additionally, the X-ray crystal structure of the sQC-LSB-09 complex revealed a unique binding mode, and a systematic computational investigation elucidated the binding mode for LSB-24. The binding mode of these two benzimidazole-6-carboxamide inhibitors offers a potential platform for designing attractive lead candidates against sQC.
Collapse
Affiliation(s)
- K V Dileep
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan; Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala 680 005, India
| | - Naoki Sakai
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Kentaro Ihara
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Akiko Nakata
- Seed Compounds Exploratory Unit for Drug Discovery Platform, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Akihiro Ito
- Seed Compounds Exploratory Unit for Drug Discovery Platform, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Laboratory of Cell Signaling, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Divya M Sivaraman
- Laboratory for Advanced Genomics Circuit, Centre for Integrative Medical Sciences, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan; Department of Pathology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695 011, Kerala, India
| | - Chi Wai Yip
- Laboratory for Advanced Genomics Circuit, Centre for Integrative Medical Sciences, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Jay W Shin
- Laboratory for Advanced Genomics Circuit, Centre for Integrative Medical Sciences, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Minoru Yoshida
- Seed Compounds Exploratory Unit for Drug Discovery Platform, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan; Office of University Professors, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Kam Y J Zhang
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.
| |
Collapse
|
109
|
Etaka JCE, Lu Y, Kang W, Salsbury FR, Derreumaux P. Impact of Amidation on Aβ 25-35 Aggregation. J Phys Chem B 2025; 129:2149-2158. [PMID: 39945395 DOI: 10.1021/acs.jpcb.4c07692] [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: 02/28/2025]
Abstract
Toxic oligomeric species are suspected in the etiology of Alzheimer's disease. The full-length Aβ42 can be studied by the fragment Aβ25-35 as it retains neurotoxicity. According to experimental studies, amidation of the Aβ25-35 carboxyl terminal decreases fibrillation activity while retaining its neurotoxic properties. Our molecular dynamics simulation studied the aggregation of the Aβ25-35 trimer from two initial structures (fibril and randomized helical structures) in their amidated and nonamidated forms. Comparing the amidated and nonamidated systems, the results suggest that antiparallel chains are dominant in nonamidated systems, while the amide group leads to parallel chains. In terms of secondary structures, a higher helix content with a corresponding decrease in β-sheet content is observed as a consequence of amidation. Despite the variation in secondary structures, the chain-chain contacts are still mediated by the Gly motif (GxxxG) and Ile residues in both amidated and nonamidated systems. As neurotoxicity does not change upon amidation, our results imply that clumping of peptides sustained by the Gly motif is a greater contributing factor to toxicity than secondary and quaternary structures.
Collapse
Affiliation(s)
- Judith C E Etaka
- School of Physics and Optoelectronic Engineering, Hainan University, Haikou 570228, China
- School of Physics, Xidian University, Xi'an 710071, China
| | - Yan Lu
- School of Physics and Optoelectronic Engineering, Hainan University, Haikou 570228, China
- School of Physics, Xidian University, Xi'an 710071, China
| | - Wei Kang
- School of Physics, Xidian University, Xi'an 710071, China
| | - Freddie R Salsbury
- Department of Physics, Wake Forest University, Winston-Salem, North Carolina 27106, United States
| | - Philippe Derreumaux
- UPR 9080 CNRS, Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, 13 Rue Pierre et Marie Curie, 75005 Paris, France
- Institut Universitaire de France (IUF) et Université Paris Cité, 75005 Paris, France
| |
Collapse
|
110
|
Zhang X, Zhang Y, Zhang T, Wang J, Liu C, Shang Q, Wei X, Zhu H, Shen H, Sun B. HCN2 deficiency correlates with memory deficits and hyperexcitability of dCA1 pyramidal neurons in Alzheimer's disease. Alzheimers Res Ther 2025; 17:55. [PMID: 40016780 PMCID: PMC11866685 DOI: 10.1186/s13195-025-01704-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Abnormal excitability of hippocampal neurons may lead to dysfunction of neural circuits and then causes cognitive impairments in Alzheimer's disease (AD). However, the underlying mechanisms remain to be fully elucidated. METHODS Electrophysiology was performed to examine the intrinsic excitability of CA1 neurons and the activity of the hyperpolarization-activated cyclic nucleotide-gated ion channels (HCNs) of CA1 neurons in wild type (WT) and hAPP-J20 mice. The activity of CA1 pyramidal neurons (PNs) was modulated with chemogenetics. The activity of HCNs was regulated with nonselective facilitator (cAMP) or inhibitor (ZD7288) of HCNs. Immunohistochemical staining or western blotting were performed to examine the expression of HCN1 and HCN2 in the hippocampus of WT and hAPP-J20 mice, or AD patients and non-AD controls. AAVs were injected to specifically modulate the expression of HCN2 in dorsal CA1 (dCA1) PNs. Cognitive performance of mice was assessed with behavioral tests. RESULTS dCA1 PNs were more excitable in hAPP-J20 mice, but the excitability of PNs in the ventral CA1 (vCA1) or PV neurons was comparable between WT and hAPP-J20 mice. The activity of the HCNs was reduced in dCA1 PNs of hAPP-J20 mice, and pharmacologically increasing the activity of HCNs attenuated the hyperexcitability of dCA1 PNs in hAPP-J20 mice, suggesting that the reduced activity of HCNs is associated with the hyperexcitability of dCA1 PNs in hAPP-J20 mice. The expression of HCN2 but not HCN1 was reduced in the hippocampus of hAPP-J20 mice, and the expression of HCN2 was also reduced in the hippocampus of AD patients, suggesting that dysregulation of HCN2 is associated with the reduced activity of HCNs in AD. Overexpressing HCN2 rescued the activity of HCNs, attenuated the hyperexcitability of dCA1 PNs and improved memory of hAPP-J20 mice, and knocking down HCN2 impaired the function of HCNs, increased the excitability of dCA1 PNs and led to memory deficits in WT mice. CONCLUSIONS Our data suggest that dysregulation of HCNs, particularly HCN2, contributes to the abnormal excitability of CA1 PNs in AD mice and probably in AD patients as well, and thus provide new insights into the mechanisms underlying the aberrant activity or excitability of hippocampal neurons in AD.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China.
| | - Yiping Zhang
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Ting Zhang
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Jing Wang
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Chang Liu
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Qing Shang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, Zhejiang Province, 315211, China
| | - Xiaojie Wei
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Huaqiang Zhu
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang Province, 315100, China
| | - Haowei Shen
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China.
| | - Binggui Sun
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China.
| |
Collapse
|
111
|
Fułek M, Hachiya N, Gachowska M, Beszłej JA, Bartoszewska E, Kurpas D, Kurpiński T, Adamska H, Poręba R, Urban S, Fułek K, Leszek J. Cellular Prion Protein and Amyloid-β Oligomers in Alzheimer's Disease-Are There Connections? Int J Mol Sci 2025; 26:2097. [PMID: 40076721 PMCID: PMC11900156 DOI: 10.3390/ijms26052097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Pathological deposits of neurotoxin proteins within the brain, such as amyloid-β and hyperphosphorylated tau tangles, are prominent features in AD. The prion protein (PrP) is involved in neurodegeneration via its conversion from the normal cellular form (PrPC) to the infection prion protein scrapie (PrPSc) form. Some studies indicated that post-translationally modified PrPC isoforms play a fundamental role in AD pathological progression. Several studies have shown that the interaction of Aβ oligomers (Aβos) with the N-terminal residues of the PrPC protein region appears critical for neuronal toxicity. PrPC-Aβ binding always occurs in AD brains and is never detected in non-demented controls, and the binding of Aβ aggregates to PrPC is restricted to the N-terminus of PrPC. In this study, we aimed to gather all of the recent information about the connections between PrPC and AD, with potential clinical implications.
Collapse
Affiliation(s)
- Michał Fułek
- Department and Clinic of Diabetology, Hypertension and Internal Diseases, Institute of Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Naomi Hachiya
- Shonan Research Center, New-STEP Research Center, Central Glass Co., Ltd., Shonan Health Innovation Park 26-1, Muraoka Higashi, Fujisawa 251-8555, Kanagawa, Japan;
| | - Martyna Gachowska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.G.); (E.B.); (T.K.)
| | - Jan Aleksander Beszłej
- Department and Clinic of Psychiatry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Elżbieta Bartoszewska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.G.); (E.B.); (T.K.)
| | - Donata Kurpas
- Division of Research Methodology, Department of Nursing, Faculty of Nursing and Midwifery, Wroclaw Medical University, 51-618 Wroclaw, Poland;
| | - Tomasz Kurpiński
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.G.); (E.B.); (T.K.)
| | - Hanna Adamska
- Department of Rheumatology and Internal Medicine, Marciniak Lower Silesian Specialist Hospital, 54-049 Wroclaw, Poland;
| | - Rafał Poręba
- Department of Biological Principles of Physical Activity, Wroclaw University of Health and Sport Sciences, 51-612 Wroclaw, Poland;
| | - Szymon Urban
- Department of Cardiology, The Copper Health Center, 59-301 Lubin, Poland;
| | - Katarzyna Fułek
- Department and Clinic of Otolaryngology, Head and Neck Surgery, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Jerzy Leszek
- Department and Clinic of Psychiatry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| |
Collapse
|
112
|
Tian Y, Torres-Flores AP, Shang Q, Zhang H, Khursheed A, Tahirbegi B, Pallier PN, Viles JH. The p3 peptides (Aβ 17-40/42) rapidly form amyloid fibrils that cross-seed with full-length Aβ. Nat Commun 2025; 16:2040. [PMID: 40016209 PMCID: PMC11868391 DOI: 10.1038/s41467-025-57341-4] [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: 06/27/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
The p3 peptides, Aβ17-40/42, are a common alternative cleavage product of the amyloid precursor protein, and are found in diffuse amyloid deposits of Alzheimer's and Down Syndrome brains. The p3 peptides have been mis-named 'non-amyloidogenic'. Here we show p340/42 peptides rapidly form amyloid fibrils, with kinetics dominated by secondary nucleation. Importantly, cross-seeding experiments, with full-length Aβ induces a strong nucleation between p3 and Aβ peptides. The cross-seeding interaction is highly specific, and occurs only when the C-terminal residues are matched. We have imaged membrane interactions with p3, and monitored Ca2+ influx and cell viability with p3 peptide. Together this data suggests the N-terminal residues influence, but are not essential for, membrane disruption. Single particle analysis of TEM images indicates p3 peptides can form ring-like annular oligomers. Patch-clamp electrophysiology, shows p342 oligomers are capable of forming large ion-channels across cellular membranes. A role for p3 peptides in disease pathology should be considered as p3 peptides are cytotoxic and cross-seed Aβ fibril formation in vitro.
Collapse
Affiliation(s)
- Yao Tian
- Department of Biochemistry, School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Andrea P Torres-Flores
- Department of Biochemistry, School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Qi Shang
- Department of Biochemistry, School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Hui Zhang
- Department of Biochemistry, School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Anum Khursheed
- Department of Biochemistry, School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Bogachan Tahirbegi
- Department of Biochemistry, School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK
| | - Patrick N Pallier
- The Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Queen Mary University of London, London, E1 2AT, UK
| | - John H Viles
- Department of Biochemistry, School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 4NS, UK.
| |
Collapse
|
113
|
Wang Y, Liu H, Ye Y, Fang W, Lin A, Dai X, Ye Q, Chen X, Zhang J. ApoE2 affects insulin signaling in the hippocampus and spatial cognition of aged mice in a sex-dependent manner. Cell Commun Signal 2025; 23:112. [PMID: 40011916 DOI: 10.1186/s12964-025-02093-3] [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: 12/20/2024] [Accepted: 02/08/2025] [Indexed: 02/28/2025] Open
Abstract
Apolipoprotein E (APOE) has garnered significant attention as one of the most influential genetic risk factors for Alzheimer's disease (AD). While the pathogenic role of APOE4 in sporadic AD has been extensively studied, research on the protective effects of the APOE2 genotype and its underlying mechanisms remains limited. Additionally, the existence of sex differences in the protective effects of ApoE2 continues to be a topic of debate. In this study, we utilized humanized ApoE2- and ApoE3- target replacement mice to examine the sex-specific effects of ApoE2 on cognition. Compared with female ApoE3 mice, we found significantly lower spatial cognitive ability and impaired hippocampal synaptic ultrastructure in aged female ApoE2 mice, accompanied by reduced insulin signaling of the hippocampus. Further analyses by target metabolomics and transcriptomic analyses revealed that female ApoE2 mice exhibit an age-related decline in hippocampal inositol levels, and that alterations in inositol levels lower insulin signaling. Importantly, inositol supplementation was found to alleviate peripheral glucose intolerance, enhance insulin signaling, and ultimately improve cognitive function. Interestingly, these differences were not observed between male ApoE2 and ApoE3 mice. The research findings not only provide new insights into the impact of ApoE2 on cognition but also offer a new strategy for cognitive improvement through inositol supplementation in older women.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Hanchen Liu
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yijuan Ye
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Wenting Fang
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Anlan Lin
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaoman Dai
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Qinyong Ye
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China.
| | - Jing Zhang
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
114
|
Li CY, Fan LY, Lin CH, Hu CJ, Chiu MJ. Ultrasensitive Assays Detect Different Conformations of Plasma β Amyloids. ACS OMEGA 2025; 10:7256-7263. [PMID: 40028141 PMCID: PMC11865983 DOI: 10.1021/acsomega.4c10879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/18/2025] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
With the developments of ultrasensitive technologies such as immunomagnetic reduction (IMR) assay, single molecule array (SIMOA) assay, electrochemiluminescence immunoassay (ECLIA), the assay of blood-based amyloid 1-42 (Aβ1-42) becomes possible. However, the changes in measured plasma Aβ1-42 concentrations in Alzheimer's disease (AD) compared to cognitively unimpaired subjects (CU) are inconsistent. A possible reason for the inconsistency regarding various conformations of Aβ1-42 in plasma is explored in this study. Three samples with equal amounts of Aβ1-42 but different proportions of monomers and oligomers of Aβ1-42 were prepared. The Aβ1-42 composition of monomers and oligomers in samples was analyzed with Western blot. Identically diluted versions of these three samples were assayed with IMR and SIMOA for Aβ1-42 concentrations. The three diluted samples showed similar levels of Aβ1-42 assayed with IMR, whereas much lower levels for samples with more oligomers assayed with SIOMA. The results imply that IMR detects both monomers and oligomers of Aβ1-42. The measured levels of Aβ1-42 are independent of the proportions of monomer or oligomer Aβ1-42 but depend on the total amounts of Aβ1-42. In the case of SIMOA, monomers of Aβ1-42 are the primary target measured. By comparing Aβ1-42 concentrations of the plasma using IMR and SIMOA, the significant difference in plasma Aβ1-42 levels using IMR in AD compared to CU is mainly due to the formations of oligomeric Aβ1-42. Therefore, if the target molecules are monomers of Aβ1-42, SIMOA is the method of choice. Still, if the target molecules should include monomers, small and large oligomers, IMR would be an optimal consideration. In the future, the clinical implications of the proportion of oligomeric Aβ1-42 need to be elucidated.
Collapse
Affiliation(s)
- Chia-Yu Li
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ling-Yun Fan
- Departments
of Neurology, National Taiwan University
Hospital Bei-Hu Branch, Taipei 108, Taiwan
| | - Chin-Hsien Lin
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Institute
of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department
of Biomedical Engineering, National Taiwan
University, Taipei 106, Taiwan
| | - Chaur-Jong Hu
- Taipei
Neuroscience Institute, Taipei Medical University, New Taipei City, 235 Taiwan
- Department
of Neurology and Dementia Center, Taipei
Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
- Department
of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Ming-Jang Chiu
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
115
|
Luo Q, Ye X, Xu J, Sun Z, Wang P, Chen H, Gao T, Deng Q, Wang Z, Wang Q, Chen X, Zhao Z, Wu Y, Yang C, Lyv W, Lyv X, Li Y, Zhao H, Jiang K, Gu Z, Lin J, Sun Y, Tan T, Xu H, Tong Z. Low-intensity pulsed ultrasound enhances delivery of 30 nm Q10 for improving mental and memory disorder in APP/PS1 mice. Drug Deliv Transl Res 2025:10.1007/s13346-025-01814-y. [PMID: 39994108 DOI: 10.1007/s13346-025-01814-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Patients with Alzheimer's disease (AD) often experience mental and memory disorders with poor outcomes. Coenzyme Q10 can degrade formaldehyde (FA) and improve Alzheimer-related symptoms, but its ability to cross the blood-brain barrier (BBB) is limited. This study investigated whether low-intensity pulsed ultrasound (LIPUS) enhances 30 nm Q10 delivery and improve symptoms in AD model mice. Here, 30 nm Q10 was prepared by encapsulating Q10 in liposomes coupled with PEG, creating PEG-Q10@NPs under 30 nm in diameter. Wild-type mice and APPswe/PS1dE9 mice (a familial AD model) received 30 nm Q10 via intraperitoneal injection, or a combination of 30 nm Q10 and LIPUS (50 or 100 100 mW/cm2). Then the mice's anxiety-like and depression-like behaviors and biochemical index were evaluated. We found that the combination therapy of LIPUS at 100 mW/cm2 and 30 nm Q10 was more effective in ameliorating psychosis in AD mice than individual treatments with 30 nm Q10. This effectiveness was linked to higher levels of brain Q10, serotonin (5-HT), and dopamine (DA), along with lower levels of FA and plaques. Especially, excessive FA directly inactivated 5-HT and DA in vitro. The enhanced cellular uptake of Q10 and improved BBB permeability facilitated by LIPUS were confirmed in both cultured cells and wild-type mice. Unexpectedly, LIPUS at the different intensity only partially alleviated anxiety and depression symptoms and memory deficits in AD mice. Hence, this combination therapy of LIPUS and 30 nm Q10 is an innovative strategy for ameliorating mental and cognitive disorders in AD.
Collapse
Affiliation(s)
- Qi Luo
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
- Beijing Geriatric Hospital, Beijing, 100095, China
| | - Xuanjie Ye
- Institute of Wenzhou, Zhejiang University, Wenzhou, 325006, China
- Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jinan Xu
- Center for Applied Psychological Research (Ningbo), School of Mental Health, Wenzhou Medical University, Cixi, 315300, China
| | - Zihui Sun
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Panpan Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Haishu Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Tingting Gao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Qiangfeng Deng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Ziwen Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Qin Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Xiaoxiao Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Zhuowen Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Yiqing Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Chuang Yang
- Department of Psychiatry, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wanjia Lyv
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
- Key Laboratory of Environmental Related Diseases and One Health, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Xingzhou Lyv
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Ye Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Hang Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Ke Jiang
- Center for Brain, Mind and Education, Shaoxing University, Shaoxing, China
| | - Ziqi Gu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Jing Lin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Yu Sun
- Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Tao Tan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.
| | - Haiyun Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.
| | - Zhiqian Tong
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Provincial Clinical Research Center for Mental Disorders, the Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.
- Beijing Geriatric Hospital, Beijing, 100095, China.
| |
Collapse
|
116
|
Atanasova M. Small-Molecule Inhibitors of Amyloid Beta: Insights from Molecular Dynamics-Part A: Endogenous Compounds and Repurposed Drugs. Pharmaceuticals (Basel) 2025; 18:306. [PMID: 40143085 PMCID: PMC11944459 DOI: 10.3390/ph18030306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
The amyloid hypothesis is the predominant model of Alzheimer's disease (AD) pathogenesis, suggesting that amyloid beta (Aβ) peptide is the primary driver of neurotoxicity and a cascade of pathological events in the central nervous system. Aβ aggregation into oligomers and deposits triggers various processes, such as vascular damage, inflammation-induced astrocyte and microglia activation, disrupted neuronal ionic homeostasis, oxidative stress, abnormal kinase and phosphatase activity, tau phosphorylation, neurofibrillary tangle formation, cognitive dysfunction, synaptic loss, cell death, and, ultimately, dementia. Molecular dynamics (MD) is a powerful structure-based drug design (SBDD) approach that aids in understanding the properties, functions, and mechanisms of action or inhibition of biomolecules. As the only method capable of simulating atomic-level internal motions, MD provides unique insights that cannot be obtained through other techniques. Integrating experimental data with MD simulations allows for a more comprehensive understanding of biological processes and molecular interactions. This review summarizes and evaluates MD studies from the past decade on small molecules, including endogenous compounds and repurposed drugs, that inhibit amyloid beta. Furthermore, it outlines key considerations for future MD simulations of amyloid inhibitors, offering a potential framework for studies aimed at elucidating the mechanisms of amyloid beta inhibition by small molecules.
Collapse
|
117
|
Trambauer J, Sarmiento RMR, Garringer HJ, Salbaum K, Pedro LD, Crusius D, Vidal R, Ghetti B, Paquet D, Baumann K, Lindemann L, Steiner H. γ-Secretase modulator resistance of an aggressive Alzheimer-causing presenilin mutant can be overcome in the heterozygous patient state by a set of advanced compounds. Alzheimers Res Ther 2025; 17:49. [PMID: 39972463 PMCID: PMC11837686 DOI: 10.1186/s13195-025-01680-3] [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: 12/11/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Amyloid-β peptide (Aβ) species of 42 or 43 amino acids in length (Aβ42/43) trigger Alzheimer´s disease (AD) and are produced in abnormal amounts by mutants of the γ-secretase subunit presenilin-1 (PS1), which represent the primary cause of familial AD (FAD). Lowering these peptides by γ-secretase modulators (GSMs) is increasingly considered a safe strategy to treat AD since these compounds do not affect the overall cleavage of γ-secretase substrates. GSMs were shown to modulate not only wild-type (WT) γ-secretase but also FAD mutants, expanding their potential use also to the familial form of the disease. Unlike most other FAD mutants, the very aggressive PS1 L166P mutant is largely resistant to GSMs. However, these data were mostly obtained from overexpression models, which mimic more the less relevant homozygous state rather than the heterozygous patient situation. METHODS Mouse embryonic fibroblast and induced pluripotent stem cell-derived neuronal PS1 L166P knock-in (KI) cell models were treated with various GSMs and Aβ responses were assessed by immunoassays and/or gel-based analysis. RESULTS We identified GSMs that lower Aβ42 and/or Aβ43 when PS1 L166P is heterozygous, as it is the case in affected patients, and could reduce the amount of pathogenic Aβ species towards WT levels. RO7019009 was the most potent of these compounds, reducing both pathogenic species and concomitantly increasing the short Aβ37 and Aβ38, of which the latter has been associated with delayed AD progression. Another effective compound, the structurally novel indole-type GSM RO5254601 specifically acts on the Aβ42 product line leading to a selective increase of the beneficial Aβ38. Interestingly, we further found that this class of GSMs can bind not only one, but both presenilin fragments suggesting that it targets γ-secretase at an unusual binding site. CONCLUSION Our data show that even highly refractory presenilin FAD mutants are in principle tractable with GSMs extending the possibilities for potential clinical studies in FAD with suitable GSM molecules.
Collapse
Affiliation(s)
- Johannes Trambauer
- Division of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU Munich, Feodor-Lynen-Str. 17, Munich, 81377, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
| | - Rosa Maria Rodriguez Sarmiento
- Pharma Research and Early Development, F. Hoffmann-La Roche AG, Therapeutic Modalities, Small Molecule Research, Roche Innovation Center Basel, Basel, 4070, Switzerland
| | - Holly J Garringer
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Katja Salbaum
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, 81377, Germany
| | - Liliana D Pedro
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, 81377, Germany
| | - Dennis Crusius
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, 81377, Germany
| | - Ruben Vidal
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Dominik Paquet
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, 81377, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, 81377, Germany
| | - Karlheinz Baumann
- Pharma Research and Early Development, F. Hoffmann-La Roche AG, Neuroscience and Rare Diseases Translational Area, Neuroscience Discovery, Roche Innovation Center Basel, Basel, 4070, Switzerland
| | - Lothar Lindemann
- Pharma Research and Early Development, F. Hoffmann-La Roche AG, Neuroscience and Rare Diseases Translational Area, Neuroscience Discovery, Roche Innovation Center Basel, Basel, 4070, Switzerland
| | - Harald Steiner
- Division of Metabolic Biochemistry, Faculty of Medicine, Biomedical Center (BMC), LMU Munich, Feodor-Lynen-Str. 17, Munich, 81377, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, 81377, Germany.
| |
Collapse
|
118
|
Naik RA, Mir MN, Malik IA, Bhardwaj R, Alshabrmi FM, Mahmoud MA, Alhomrani M, Alamri AS, Alsanie WF, Hjazi A, Ghatak T, Poeggeler B, Singh MP, Ts G, Singh SK. The Potential Mechanism and the Role of Antioxidants in Mitigating Oxidative Stress in Alzheimer's Disease. FRONT BIOSCI-LANDMRK 2025; 30:25551. [PMID: 40018917 DOI: 10.31083/fbl25551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/30/2024] [Accepted: 08/23/2024] [Indexed: 03/01/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia and a significant contributor to health issues and mortality among older individuals. This condition involves a progressive deterioration in cognitive function and the onset of dementia. Recent advancements suggest that the development of AD is more intricate than its underlying brain abnormalities alone. In addition, Alzheimer's disease, metabolic syndrome, and oxidative stress are all intricately linked to one another. Increased concentrations of circulating lipids and disturbances in glucose homeostasis contribute to the intensification of lipid oxidation, leading to a gradual depletion of the body's antioxidant defenses. This heightened oxidative metabolism adversely impacts cell integrity, resulting in neuronal damage. Pathways commonly acknowledged as contributors to AD pathogenesis include alterations in synaptic plasticity, disorganization of neurons, and cell death. Abnormal metabolism of some membrane proteins is thought to cause the creation of amyloid (Aβ) oligomers, which are extremely hazardous to neurotransmission pathways, especially those involving acetylcholine. The interaction between Aβ oligomers and these neurotransmitter systems is thought to induce cellular dysfunction, an imbalance in neurotransmitter signaling, and, ultimately, the manifestation of neurological symptoms. Antioxidants have a significant impact on human health since they may improve the aging process by combating free radicals. Neurodegenerative diseases are currently incurable; however, they may be effectively managed. An appealing alternative is the utilization of natural antioxidants, such as polyphenols, through diet or dietary supplements, which offer numerous advantages. Within this framework, we have extensively examined the importance of oxidative stress in the advancement of Alzheimer's disease, as well as the potential influence of antioxidants in mitigating its effects.
Collapse
Affiliation(s)
- Rayees Ahmad Naik
- Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya Sagar, 470003 Sagar, Madhya Pradesh, India
| | - Mehak Naseer Mir
- NIMS Institute of Allied Medical Science, National Institute of Medical Sciences (NIMS), 303121 Jaipur, Rajasthan, India
| | - Ishfaq Ahmad Malik
- Department of Zoology, Bar. Ramrao Deshmukh Arts, Smt. Indiraji Kapadia Commerce & Nya. Krishnarao Deshmukh Science College, 444701 Amravati, Maharashtra, India
| | - Rima Bhardwaj
- Department of Chemistry Poona College, Savitribai Phule Pune University, 411007 Pune, Maharashtra, India
| | - Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Mahmoud Abdulrahman Mahmoud
- Department of Family & Community Medicine, College of Medicine, Imam Muhammad Ibn Saud Islamic University, 13313 Riyadh, Saudi Arabia
| | - Majid Alhomrani
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, 21944 Taif, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, 21944 Taif, Saudi Arabia
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, 21944 Taif, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, 21944 Taif, Saudi Arabia
| | - Walaa F Alsanie
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, 21944 Taif, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, 21944 Taif, Saudi Arabia
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, 11942 Al-Kharj, Saudi Arabia
| | - Tanmoy Ghatak
- Department of Emergency Medicine, Sanjay Gandhi Post Graduate Institute of Medical Sciences, 226014 Lucknow, Uttar Pradesh, India
| | - Burkhard Poeggeler
- Department of Physiology, Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, Faculty of Biology Georg August University Göttingen, Göttingen and Goettingen Research Campus, D-38524 Sassenburg, Germany
| | - Mahendra P Singh
- Department of Zoology, Deen Dayal Upadhyaya Gorakhpur University, 273009 Gorakhpur, Uttar Pradesh, India
| | - Gopenath Ts
- Department of Biotechnology & Bioinformatics, JSS Academy of Higher Education & Research, 570015 Mysuru, Karnataka, India
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, 226001 Lucknow, Uttar Pradesh, India
| |
Collapse
|
119
|
Lindberg M, Hu J, Sparr E, Linse S. Reduced protein solubility - cause or consequence in amyloid disease? QRB DISCOVERY 2025; 6:e8. [PMID: 40070848 PMCID: PMC11894405 DOI: 10.1017/qrd.2024.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 03/14/2025] Open
Abstract
In this perspective, we ask the question whether the apparently lower solubility of specific proteins in amyloid disease is a cause or consequence of the protein deposition seen in such diseases. We focus on Alzheimer's disease and start by reviewing the experimental evidence of disease-associated reduction in the measured concentration of amyloid β peptide, Aβ42, in cerebrospinal fluid. We propose a series of possible physicochemical explanations for these observations. These include a reduced solubility, a reduced apparent solubility, as well as a long-lived metastable state manifested in healthy individuals as a free concentration of Aβ42 in the solution phase above the solubility limit. For each scenario, we discuss whether it is most likely a cause or a consequence of the observed protein deposition in the disease.
Collapse
Affiliation(s)
- Max Lindberg
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Jing Hu
- Division for Physical Chemistry, Lund University, Lund, Sweden
| | - Emma Sparr
- Division for Physical Chemistry, Lund University, Lund, Sweden
| | - Sara Linse
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| |
Collapse
|
120
|
Feng YY, Hao JR, Zhang YJ, Qiu TT, Zhang ML, Qiao W, Wu JJ, Qiu P, Xu CF, Zhang YL, Du CY, Pan Z, Chang YS. Krüppel-like factor 9 alleviates Alzheimer's disease via IDE-mediated Aβ degradation. Acta Pharmacol Sin 2025:10.1038/s41401-025-01491-0. [PMID: 39962264 DOI: 10.1038/s41401-025-01491-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/19/2025] [Indexed: 03/17/2025]
Abstract
The deposition of β-amyloid (Aβ) in the brain is a crucial factor in the pathogenesis of Alzheimer's disease (AD). Insulin-degrading enzyme (IDE) plays a critical role in the balance between Aβ production and degradation. However, the regulatory mechanisms of IDE are not yet fully understood. Therefore, uncovering additional IDE regulatory mechanisms will help elucidate the pathogenesis of AD and identify key therapeutic targets for this disease. This study revealed that global Krüppel-like factor 9-mutant (Klf9-/-) mice exhibited impaired cognitive function. Additionally, we found that Klf9 expression in hippocampal tissue was reduced in APPswe/PS1dE9 (APP/PS1) mice. This study also showed that Klf9 stimulates IDE expression and promotes the Aβ degradation process by directly binding to IDE and activating its transcription. Silencing IDE blocked the Klf9-induced Aβ degradation process. We stereotactically injected an adeno-associated virus to selectively overexpress IDE (AAV-IDE) in the hippocampal neurons of Klf9-/- mice and found that the overexpression of IDE in hippocampal neurons ameliorated cognitive deficits and reduced the Aβ content in Klf9-/- mice. Additionally, we also stereotactically injected AAV-Klf9 into the hippocampal neurons of APP/PS1 mice and found that overexpression of Klf9 in hippocampal neurons ameliorated cognitive deficits and reduced Aβ levels in APP/PS1 mice. These findings suggest that downregulation of Klf9 may be a key factor in AD progression, as it reduces Aβ clearance by decreasing IDE expression. Overexpression or activation of Klf9 may be a potential strategy for preventing the pathogenesis of AD.
Collapse
Affiliation(s)
- Yue-Yao Feng
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Jing-Ran Hao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Yu-Jie Zhang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Tong-Tong Qiu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Meng-Lin Zhang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Wei Qiao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Jin-Jin Wu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Ping Qiu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Chao-Fan Xu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Yin-Liang Zhang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Chun-Yuan Du
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China
| | - Zhe Pan
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, 250033, China.
| | - Yong-Sheng Chang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300052, China.
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300052, China.
| |
Collapse
|
121
|
Spooner A, Mohammadi G, Sachdev PS, Brodaty H, Sowmya A. Identifying risk factors for Alzheimer's disease from multivariate longitudinal clinical data using temporal pattern mining. BMC Bioinformatics 2025; 26:56. [PMID: 39962385 PMCID: PMC11834509 DOI: 10.1186/s12859-024-06018-8] [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] [Accepted: 12/17/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Patient data contain a wealth of information that could aid in understanding the onset and progression of disease. However, the task of modelling clinical data, which consist of multiple heterogeneous time series of different lengths, measured at different time intervals, is a complex one. A growing body of research has applied temporal pattern mining to this problem to identify common patterns in clinical attributes over time. However, the vast majority of these algorithms use techniques that are not ideally suited to clinical data. We present an efficient and scalable framework designed specifically for temporal pattern mining of real-world clinical data. Our framework combines temporal abstraction, an extended version of the efficient pattern-growth algorithm, TPMiner, the concepts of relative risk and the odds ratio to identify interesting and high-risk patterns and multiprocessing to improve computational efficiency. A complete set of cut-off values for discretisation and interpretation of the data is provided and is applicable to studies on ageing populations in general. We name this framework Clinical Temporal Pattern Mining or C-TPM. RESULTS The framework is applied to data from two real-world studies of Alzheimer's disease (AD). The patterns discovered were predictive of AD in survival analysis models with a Concordance index of up to 0.87 and contain clinically relevant variables. A visualisation module provides a clear picture of the discovered patterns for ease of interpretability. CONCLUSIONS The framework provides an effective and scalable method of modelling multivariate, longitudinal clinical data and can identify patterns in uncommon diseases and those that progress slowly over time. It is generalisable to clinical data from other medical domains as well as non-clinical data.
Collapse
Affiliation(s)
- Annette Spooner
- School of Computer Science and Engineering, UNSW Sydney, Sydney, Australia.
| | - Gelareh Mohammadi
- School of Computer Science and Engineering, UNSW Sydney, Sydney, Australia
| | - Perminder S Sachdev
- Faculty of Medicine and Health, Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, UNSW Sydney, Sydney, Australia
| | - Henry Brodaty
- Faculty of Medicine and Health, Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, UNSW Sydney, Sydney, Australia
| | - Arcot Sowmya
- School of Computer Science and Engineering, UNSW Sydney, Sydney, Australia
| |
Collapse
|
122
|
Patel R, Cheng R, Cardona CL, Angeles E, Singh G, Miller S, Ashok A, Teich AF, Piriz A, Maldonado A, Jimenez-Velazquez IZ, Mayeux R, Lee JH, Sproul AA. Reduced SH3RF3 may protect against Alzheimer's disease by lowering microglial pro-inflammatory responses via modulation of JNK and NFkB signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.23.600281. [PMID: 38979369 PMCID: PMC11230201 DOI: 10.1101/2024.06.23.600281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Understanding how high-risk individuals are protected from Alzheimer's disease (AD) may illuminate potential therapeutic targets. We identified protective genetic variants in SH3RF3/POSH2 that delayed the onset of AD among individuals carrying the PSEN1 G206A mutation. SH3RF3 acts as a JNK pathway scaffold and activates NFκB signaling. While effects of SH3RF3 knockdown in human neurons were subtle, including decreased ptau S422, knockdown in human microglia significantly reduced inflammatory cytokines in response to either a viral mimic or oAβ42. This was associated with reduced activation of JNK and NFκB pathways in response to these stimuli. Pharmacological inhibition of JNK or NFκB signaling phenocopied SH3RF3 knockdown. We also found PSEN1 G206A microglia had reduced inflammatory response to oAβ42. Thus, further reduction of microglial inflammatory responses in PSEN1 G206A mutant carriers by protective variants in SH3RF3 might reduce the link between amyloid and neuroinflammation to subsequently delay the onset of AD.
Collapse
|
123
|
Paitel ER, Otteman CBD, Polking MC, Licht HJ, Nielson KA. Functional and effective EEG connectivity patterns in Alzheimer's disease and mild cognitive impairment: a systematic review. Front Aging Neurosci 2025; 17:1496235. [PMID: 40013094 PMCID: PMC11861106 DOI: 10.3389/fnagi.2025.1496235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025] Open
Abstract
Background Alzheimer's disease (AD) might be best conceptualized as a disconnection syndrome, such that symptoms may be largely attributable to disrupted communication between brain regions, rather than to deterioration within discrete systems. EEG is uniquely capable of directly and non-invasively measuring neural activity with precise temporal resolution; connectivity quantifies the relationships between such signals in different brain regions. EEG research on connectivity in AD and mild cognitive impairment (MCI), often considered a prodromal phase of AD, has produced mixed results and has yet to be synthesized for comprehensive review. Thus, we performed a systematic review of EEG connectivity in MCI and AD participants compared with cognitively healthy older adult controls. Methods We searched PsycINFO, PubMed, and Web of Science for peer-reviewed studies in English on EEG, connectivity, and MCI/AD relative to controls. Of 1,344 initial matches, 124 articles were ultimately included in the systematic review. Results The included studies primarily analyzed coherence, phase-locked, and graph theory metrics. The influence of factors such as demographics, design, and approach was integrated and discussed. An overarching pattern emerged of lower connectivity in both MCI and AD compared to healthy controls, which was most prominent in the alpha band, and most consistent in AD. In the minority of studies reporting greater connectivity, theta band was most commonly implicated in both AD and MCI, followed by alpha. The overall prevalence of alpha effects may indicate its potential to provide insight into nuanced changes associated with AD-related networks, with the caveat that most studies were during the resting state where alpha is the dominant frequency. When greater connectivity was reported in MCI, it was primarily during task engagement, suggesting compensatory resources may be employed. In AD, greater connectivity was most common during rest, suggesting compensatory resources during task engagement may already be exhausted. Conclusion The review highlighted EEG connectivity as a powerful tool to advance understanding of AD-related changes in brain communication. We address the need for including demographic and methodological details, using source space connectivity, and extending this work to cognitively healthy older adults with AD risk toward advancing early AD detection and intervention.
Collapse
Affiliation(s)
- Elizabeth R. Paitel
- Aging, Imaging, and Memory Laboratory, Department of Psychology, Marquette University, Milwaukee, WI, United States
| | - Christian B. D. Otteman
- Aging, Imaging, and Memory Laboratory, Department of Psychology, Marquette University, Milwaukee, WI, United States
| | - Mary C. Polking
- Aging, Imaging, and Memory Laboratory, Department of Psychology, Marquette University, Milwaukee, WI, United States
| | - Henry J. Licht
- Aging, Imaging, and Memory Laboratory, Department of Psychology, Marquette University, Milwaukee, WI, United States
| | - Kristy A. Nielson
- Aging, Imaging, and Memory Laboratory, Department of Psychology, Marquette University, Milwaukee, WI, United States
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
124
|
Futácsi A, Rusznák K, Szarka G, Völgyi B, Wiborg O, Czéh B. Quantification and correlation of amyloid-β plaque load, glial activation, GABAergic interneuron numbers, and cognitive decline in the young TgF344-AD rat model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1542229. [PMID: 40013092 PMCID: PMC11860898 DOI: 10.3389/fnagi.2025.1542229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025] Open
Abstract
Background Animal models of Alzheimer's disease (AD) are essential tools for investigating disease pathophysiology and conducting preclinical drug testing. In this study, we examined neuronal and glial alterations in the hippocampus and medial prefrontal cortex (mPFC) of young TgF344-AD rats and correlated these changes with cognitive decline and amyloid-β plaque load. Methods We compared TgF344-AD and non-transgenic littermate rats aged 7-8 months of age. We systematically quantified β-amyloid plaques, astrocytes, microglia, four different subtypes of GABAergic interneurons (calretinin-, cholecystokinin-, parvalbumin-, and somatostatin-positive neurons), and newly generated neurons in the hippocampus. Spatial learning and memory were assessed using the Barnes maze test. Results Young TgF344-AD rats had a large number of amyloid plaques in both the hippocampus and mPFC, together with a pronounced increase in microglial cell numbers. Astrocytic activation was significant in the mPFC. Cholecystokinin-positive cell numbers were decreased in the hippocampus of transgenic rats, but calretinin-, parvalbumin-, and somatostatin-positive cell numbers were not altered. Adult neurogenesis was not affected by genotype. TgF344-AD rats had spatial learning and memory impairments, but this cognitive deficit did not correlate with amyloid plaque number or cellular changes in the brain. In the hippocampus, amyloid plaque numbers were negatively correlated with cholecystokinin-positive neuron and microglial cell numbers. In the mPFC, amyloid plaque number was negatively correlated with the number of astrocytes. Conclusion Pronounced neuropathological changes were found in the hippocampus and mPFC of young TgF344-AD rats, including the loss of hippocampal cholecystokinin-positive interneurons. Some of these neuropathological changes were negatively correlated with amyloid-β plaque load, but not with cognitive impairment.
Collapse
Affiliation(s)
- Anett Futácsi
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
- Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Kitti Rusznák
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Gergely Szarka
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Neurobiology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Neurobiology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Ove Wiborg
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Boldizsár Czéh
- Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
- Imaging Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
125
|
Qu T. The effects of amyloidosis and aging on glutamatergic and GABAergic synapses, and interneurons in the barrel cortex and non-neocortical brain regions. Front Neuroanat 2025; 19:1526962. [PMID: 40012738 PMCID: PMC11863279 DOI: 10.3389/fnana.2025.1526962] [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: 11/12/2024] [Accepted: 01/15/2025] [Indexed: 02/28/2025] Open
Abstract
Previous studies on changes in the distribution of GABAergic interneurons and excitation/inhibition (E/I) balance in Alzheimer's disease (AD) and aging were mainly conducted in the neocortex and hippocampus. However, the limbic system is the primary and crucial location for AD progression. Therefore, in this study, we utilized AD and aging mouse models to investigate the E/I balance and the distribution of parvalbumin (PV)- and somatostatin (SST)-expressing cells in S1BF (barrel field of primary somatosensory cortex, barrel cortex), CA1 hippocampal area and brain regions beyond the neocortex and hippocampus, including retrosplenial cortex (RSC, which is composed of RSG and RSA), piriform cortex (Pir), amygdala (BMA), and hypothalamus (DM). We discovered that amyloidosis may disrupt the alignment of excitatory pre- and postsynaptic quantities. Amyloidosis reduces the quantity of synapses and SST cells, but does not impact the counts of PV cells. By contrast, aging is linked to a decline in synapses, I/E ratios, SST and PV cells. Amyloidosis affects the S1BF and BMA, while aging may harm all studied regions, including the S1BF, RSC, hippocampus, Pir, BMA, and DM. Aging mostly affects synapses and I/E ratios in Pir, BMA, and DM, and PV and SST interneurons in the hippocampus.
Collapse
Affiliation(s)
- Tao Qu
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
126
|
Iida-Adachi A, Nabika H. Changes in Adsorption, Aggregation, and Diffusion Nature of Amyloid β on a Lipid Membrane in an Open System. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:3121-3129. [PMID: 39883914 DOI: 10.1021/acs.langmuir.4c03663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
The aggregation and accumulation of amyloid β 42 (Aβ42) peptides on the surface of brain cells is associated with Alzheimer's disease (AD); however, the underlying molecular mechanisms remain unclear. Herein, we used a unique brain-mimetic open system that continuously flows Aβ42 solution to analyze the initial aggregation and adsorptive nature of Aβ42 at physiological concentrations on the lipid membrane. The open system accelerated the adsorption and dimerization kinetics. Upon the addition of Aβ42, monomeric Aβ42 was dominant on the lipid bilayer surface in the closed system with no flow, whereas dimers and high-order oligomers were dominant in the open system. Closed and open systems exhibited different oligomerization kinetics, lipid-Aβ42 interactions, and diffusive properties of monomers and oligomers. These results indicate the specific adsorptive and diffusive nature of Aβ42 on the cell membrane. The open system may help in elucidating the molecular mechanisms underlying AD progression.
Collapse
Affiliation(s)
- Akane Iida-Adachi
- Department of Science, Graduate School of Science and Engineering, Yamagata University, 1-4-12, Kojirakawa, Yamagata 990-8560, Japan
| | - Hideki Nabika
- Faculty of Science, Yamagata University, 1-4-12, Kojirakawa, Yamagata 990-8560, Japan
| |
Collapse
|
127
|
Arafi P, Devkota S, Williams E, Maesako M, Wolfe MS. Alzheimer-mutant γ-secretase complexes stall amyloid β-peptide production. eLife 2025; 13:RP102274. [PMID: 39932776 PMCID: PMC11813224 DOI: 10.7554/elife.102274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
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 Caenorhabditis 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 to 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 KansasLawrenceUnited States
| | - Sujan Devkota
- Department of Medicinal Chemistry, University of KansasLawrenceUnited States
| | - Emily Williams
- Alzheimer Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
| | - Masato Maesako
- Alzheimer Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
| | - Michael S Wolfe
- Department of Medicinal Chemistry, University of KansasLawrenceUnited States
| |
Collapse
|
128
|
Sidsworth D, Tregobov N, Jamieson C, Reutens-Hernandez J, Yoon J, Payne GW, Sellers SL. Microvascular dysfunction in a murine model of Alzheimer's disease using intravital microscopy. Front Aging Neurosci 2025; 17:1482250. [PMID: 39995945 PMCID: PMC11848520 DOI: 10.3389/fnagi.2025.1482250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 01/13/2025] [Indexed: 02/26/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurocognitive disorder. Early theories of AD sought to identify a single unifying explanation underlying AD pathogenesis; however, evolving evidence suggests it is a multifactorial, systemic disease, involving multiple systems. Of note, vascular dysfunction, encompassing both cerebral and peripheral circulation, has been implicated in AD pathogenesis. This pilot study used intravital microscopy to assess differences in responsiveness of gluteal muscle arterioles between a transgenic AD mouse model (APP/PS1; Tg) and wild-type (C57BL/6; WT) mice to further elucidate the role of vascular dysfunction in AD. Arteriole diameters were measured in response to acetylcholine (10-9 to 10-5 M), phenylephrine (10-9 to 10-5 M), histamine (10-9 to 10-4 M) and compound 48/80 (10-9 to 10-3 M). Tg mice demonstrated a trend toward reduced vasodilatory response to acetylcholine with a significant difference at 10-5 M (36.91 vs. 69.55%: p = 0.0107) when compared to WT. No significant differences were observed with histamine, compound 48/80 or phenylephrine; however, a trend toward reduced vasoconstriction to phenylephrine was observed in Tg mice at higher concentrations. Mean net diameter change (resting to maximum) also differed significantly (p = 0.0365) between WT (19.11 μm) and Tg mice (11.13 μm). These findings suggest reduced vascular responsiveness may contribute to the systemic vascular deficits previously observed in AD models. Future research using diverse models and broader variables could further elucidate peripheral vascular dysfunction's role in AD pathogenesis, including its impact on motor symptoms and disease progression. Such insights may inform the development of vascular-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Danielle Sidsworth
- Division of Medical Sciences, University of Northern British Columbia, Prince George, BC, Canada
| | - Noah Tregobov
- Department of Radiology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Cardiovascular Translational Laboratory, Providence Research and Centre for Heart Lung Innovation, Vancouver, BC, Canada
| | - Colin Jamieson
- Cardiovascular Translational Laboratory, Providence Research and Centre for Heart Lung Innovation, Vancouver, BC, Canada
| | - Jennifer Reutens-Hernandez
- Biochemistry and Molecular Biology Program, University of Northern British Columbia, Prince George, BC, Canada
| | - Joshua Yoon
- Cardiovascular Translational Laboratory, Providence Research and Centre for Heart Lung Innovation, Vancouver, BC, Canada
| | - Geoffrey W. Payne
- Division of Medical Sciences, University of Northern British Columbia, Prince George, BC, Canada
- University of Northern British Columbia, Prince George, BC, Canada
| | - Stephanie L. Sellers
- Cardiovascular Translational Laboratory, Providence Research and Centre for Heart Lung Innovation, Vancouver, BC, Canada
- Centre for Cardiovascular Innovation, St. Paul’s and Vancouver General Hospital, Vancouver, BC, Canada
- Centre for Heart Valve Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- Dilawri Cardiovascular Institute, Vancouver General Hospital, Vancouver, BC, Canada
| |
Collapse
|
129
|
Zhou W, Wang C, Tan Y, Lazarovici P, Wen X, Li S, Zheng W. Cordycepin mediates neuroprotection against apoptosis via ERK/CREB signaling activation in Aβ 1-42-induced neuronal cell models. IBRAIN 2025; 11:84-97. [PMID: 40103703 PMCID: PMC11911103 DOI: 10.1002/ibra.12192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 12/10/2024] [Accepted: 01/06/2025] [Indexed: 03/20/2025]
Abstract
The aggregation of β-amyloid (Aβ) peptides has been associated with the onset of Alzheimer's disease (AD) by causing neurotoxicity due to oxidative stress and apoptosis. Cordycepin is a natural derivative of the nucleoside adenosine that displays potent antioxidant, antitumor, anti-inflammatory, and neuroprotective properties. However, the mechanism of the neuroprotective effect of cordycepin toward Aβ-induced neurotoxicity, as well as underlying mechanisms, is still unclear. In this study, we found that cordycepin conferred neuroprotection to catecholaminergic PC12 neuronal cell cultures exposed to Aβ1-42-insult by reducing the production of reactive oxygen species, restoring the mitochondrial membrane potential, and inhibiting apoptosis. Cordycepin stimulated the phosphorylation of extracellular signal-regulated kinase (ERK) and cyclic AMP-responsive element-binding protein (CREB) in a time- and concentration-dependent manner. Inhibition of the ERK pathway reduced the neuroprotective effect of cordycepin. Similar results were obtained with hippocampal HT22 neuronal cell cultures. Cumulatively, these findings suggest that cordycepin-induced neuroprotection toward Aβ1-42 neurotoxic insult may involve activation of the ERK/CREB pathway. This study expands our knowledge of the neuroprotective function of cordycepin and suggests that it holds promise as a natural lead compound for drug development in AD.
Collapse
Affiliation(s)
- Wenshu Zhou
- Faculty of Health Sciences, and Zhuhai UM Science & Technology Research Institute University of Macau Macau SAR China
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine Kunming University of Science and Technology Kunming China
| | - Cheng Wang
- State Key Laboratory for Quality Research in Chinese Medicine University of Macau Macao SAR China
| | - Yige Tan
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine Kunming University of Science and Technology Kunming China
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine The Hebrew University of Jerusalem Jerusalem Israel
| | - Xiaoyan Wen
- Zhongshan Key Laboratory of Zebrafish-based Drug Innovation, ZebraPeutics (Zhongshan) Ltd. Zhongshan China
| | - Shaoping Li
- State Key Laboratory for Quality Research in Chinese Medicine University of Macau Macao SAR China
| | - Wenhua Zheng
- Faculty of Health Sciences, and Zhuhai UM Science & Technology Research Institute University of Macau Macau SAR China
| |
Collapse
|
130
|
Liu Y, Xu X, Wu X, Yang G, Luo J, Liang X, Chen J, Li Y. TMF Attenuates Cognitive Impairment and Neuroinflammation by Inhibiting the MAPK/NF-κB Pathway in Alzheimer's Disease: A Multi-Omics Analysis. Mar Drugs 2025; 23:74. [PMID: 39997198 PMCID: PMC11857128 DOI: 10.3390/md23020074] [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: 01/09/2025] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
The rising prevalence of Alzheimer's disease (AD) underscores the urgent need for novel therapeutic agents derived from natural sources. Among flavonoids, 3',4',5,7-tetramethoxyflavone (TMF), a structural analog of luteolin, has gained attention for its favorable pharmacokinetics and potential neuroprotective properties. Despite the significant neuroprotective effects and favorable pharmacokinetics of TMF, its efficacy and mechanism of action in AD remain unclear. This study explored TMF's pharmacological effects in AD models, highlighting its ability to improve memory and cognitive deficits in APP/PS1 mice. TMF reduced Aβ plaques, NFTs formation, and glial activation while suppressing neuroinflammation through the MAPK/NF-κB pathway. Further analysis in LPS-induced BV2 cells revealed TMF's ability to reduce microglial activation. These findings highlight the anti-neuroinflammatory activity of TMF, suggesting its potential as a treatment for AD.
Collapse
Affiliation(s)
- Yonglin Liu
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang 330096, China; (Y.L.); (X.L.)
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang 330103, China
| | - Xi Xu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
| | - Xiaoming Wu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
| | - Guodong Yang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
| | - Jiaxin Luo
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
| | - Xinli Liang
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang 330096, China; (Y.L.); (X.L.)
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang 330103, China
| | - Jie Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
| | - Yiguang Li
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang 330096, China; (Y.L.); (X.L.)
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang 330103, China
| |
Collapse
|
131
|
Liu L, He H, Du B, He Y. Nanoscale drug formulations for the treatment of Alzheimer's disease progression. RSC Adv 2025; 15:4031-4078. [PMID: 39926227 PMCID: PMC11803502 DOI: 10.1039/d4ra08128e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder with no effective disease-modifying treatments. The blood-brain barrier hinders drug delivery to the brain, limiting therapeutic efficacy. Nanoparticle-based systems have emerged as promising tools to overcome these challenges. This review highlights recent advances in nanoparticle technologies for AD treatment, including liposomes, polymeric, inorganic, and biomimetic nanoparticles. These nanoparticles improve drug delivery across the blood-brain barrier, improve stability and bioavailability, and enable targeted delivery to affected brain regions. Functionalization strategies further enhance their therapeutic potential. Multifunctional nanoparticles combining therapeutic and diagnostic properties offer theranostic approaches. While progress has been made, challenges related to safety, targeting precision, and clinical translation remain. Future perspectives emphasize the need for collaborative efforts to optimize nanoparticle design, conduct rigorous studies, and accelerate the development of effective nanotherapeutics. With continued innovation, nanoparticle-based delivery systems hold great promise for revolutionizing AD treatment.
Collapse
Affiliation(s)
- Liqin Liu
- Department of Pediatrics of Neurology Nursing, West China School of Nursing, West China Second University Hospital, Sichuan University Chengdu 610000 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu 610000 China
| | - Haini He
- Department of Pediatrics of Neurology Nursing, West China School of Nursing, West China Second University Hospital, Sichuan University Chengdu 610000 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu 610000 China
| | - Bin Du
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University Chengdu 610000 China
| | - Yang He
- Department of Pediatrics, West China Second University Hospital, Sichuan University Chengdu 610000 China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education Chengdu 610000 China
| |
Collapse
|
132
|
Fischer L, Molloy EN, Pichet Binette A, Vockert N, Marquardt J, Pacha Pilar A, Kreissl MC, Remz J, Tremblay-Mercier J, Poirier J, Rajah MN, Villeneuve S, Maass A. Precuneus Activity during Retrieval Is Positively Associated with Amyloid Burden in Cognitively Normal Older APOE4 Carriers. J Neurosci 2025; 45:e1408242024. [PMID: 39788739 PMCID: PMC11800745 DOI: 10.1523/jneurosci.1408-24.2024] [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: 07/23/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025] Open
Abstract
The precuneus is a site of early amyloid-beta (Aβ) accumulation. Previous cross-sectional studies reported increased precuneus fMRI activity in older adults with mild cognitive deficits or elevated Aβ. However, longitudinal studies in early Alzheimer's disease (AD) are lacking and the relationship to the Apolipoprotein-E (APOE) genotype is unclear. Investigating the PREVENT-AD dataset, we assessed how baseline and longitudinal precuneus activity during successful memory retrieval relates to future Aβ and tau burden and change in memory performance. We further studied the moderation by APOE4 genotype. We included 165 older adults (age, 62.8 ± 4.4 years; 113 female; 66 APOE4 carriers) who were cognitively normal at baseline with a family history of AD. All participants performed task-fMRI at baseline and underwent 18F-flortaucipir-PET and 18F-NAV4694-Aβ-PET on average 5 years later. We found that higher baseline activity and greater longitudinal increase in precuneus activity were associated with higher Aβ burden in APOE4 carriers but not noncarriers. We observed no effects of precuneus activity on tau burden. Finally, APOE4 noncarriers with low baseline precuneus activity exhibited better longitudinal performance in an independent memory test compared with (1) noncarriers with higher baseline activity and (2) APOE4 carriers. Our findings suggest that higher task-related precuneus activity during memory retrieval at baseline and over time are associated with greater Aβ burden in cognitively normal APOE4 carriers. Our results further indicate that the absence of "hyperactivation" and the absence of the APOE4 allele is related with better future cognitive outcomes in cognitively normal older adults at risk for AD.
Collapse
Affiliation(s)
- Larissa Fischer
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
| | - Eóin N Molloy
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
- Division of Nuclear Medicine, Department of Radiology & Nuclear Medicine, Faculty of Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany
| | - Alexa Pichet Binette
- Clinical Memory Research, Faculty of Medicine, Lund University, Lund 223 62, Sweden
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
| | - Niklas Vockert
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
| | - Jonas Marquardt
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
| | - Andrea Pacha Pilar
- Institute for Biology, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany
| | - Michael C Kreissl
- Division of Nuclear Medicine, Department of Radiology & Nuclear Medicine, Faculty of Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany
| | - Jordana Remz
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
| | - Jennifer Tremblay-Mercier
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montréal H3A 1A1, Canada
| | - Maria Natasha Rajah
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montréal H3A 1A1, Canada
- Department of Psychology, Toronto Metropolitan University, Toronto M5S 1A1, Canada
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montréal H3A 1A1, Canada
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
- Institute for Biology, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany
| |
Collapse
|
133
|
Zheng H, Mizokami A, Romera-Giner S, Llera-Oyola J, Yamawaki Y, Sano T, Jimi E, García-García F, Kanematsu T. Sex differences in the neuroinflammatory signaling pathway: effect of miRNAs on fatty acid synthesis in microglia. Biol Sex Differ 2025; 16:9. [PMID: 39905477 PMCID: PMC11792555 DOI: 10.1186/s13293-025-00686-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/13/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Significant sex differences exist in the prevalence and incidence of Alzheimer's disease (AD). Notably, testosterone has been reported to regulate cognitive functions in the brain, with low serum testosterone levels correlating with increased AD risk. However, the specific mechanisms underlying this relationship remain unclear. Recent studies have demonstrated that microglia, the primary innate immune cells in the brain, play a crucial role in AD development. Therefore, this study aimed to explore sex differences in microglial function, specifically focusing on the role of testosterone in miRNA-mediated regulation of microglial gene expression. METHODS Microglia were isolated from pooled hippocampal tissue of five 8-month-old male and female mice. Total RNA was extracted and subjected to miRNA microarray analysis. The mouse microglial cell line MG6 was used for in vitro experiments. Following testosterone treatment, miRNA, gene, and protein expression levels were investigated. An inflammatory response was induced using lipopolysaccharide (LPS) stimulation, and subsequent p65 phosphorylation was assessed. RESULTS Sex-dependent differences were observed in miRNA-mediated biological processes, with males exhibiting greater changes. Male-enriched miRNAs were associated with fatty acid synthesis and metabolism pathways. In MG6 cells, testosterone treatment upregulated the expression of several miRNAs enriched in male microglia, particularly those targeting genes related to fatty acid synthesis. Additionally, testosterone significantly reduced the gene expression of fatty acid synthase (FASN). This testosterone-induced inhibition of FASN expression attenuated NF-κB/p65 phosphorylation. Consequently, the suppression of FASN expression led to reduced expression and secretion of tumor necrosis factor-alpha following LPS stimulation in MG6 cells. CONCLUSIONS These findings suggest that testosterone modulates inflammation in male microglia by regulating fatty acid synthesis, potentially contributing to the observed sex differences in AD pathogenesis.
Collapse
Affiliation(s)
- Haolin Zheng
- Department of Cell Biology, Aging Science, and Pharmacology, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Akiko Mizokami
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| | - Sergio Romera-Giner
- Computational Biomedicine Laboratory, Prince Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Jaime Llera-Oyola
- Computational Biomedicine Laboratory, Prince Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Yosuke Yamawaki
- Department of Advanced Pharmacology, Daiichi University of Pharmacy, 22-1 Tamagawa-Cho, Minami-Ku, Fukuoka, 815-8511, Japan
| | - Tomomi Sano
- Department of Cell Biology, Aging Science, and Pharmacology, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Eijiro Jimi
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Francisco García-García
- Computational Biomedicine Laboratory, Prince Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Takashi Kanematsu
- Department of Cell Biology, Aging Science, and Pharmacology, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
134
|
Kovacs GG, Katsumata Y, Wu X, Aung KZ, Fardo DW, Forrest SL, Nelson PT. Amyloid-β predominant Alzheimer's disease neuropathologic change. Brain 2025; 148:401-407. [PMID: 39417691 PMCID: PMC11788189 DOI: 10.1093/brain/awae325] [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: 07/24/2024] [Revised: 08/22/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Different subsets of Alzheimer's disease neuropathologic change (ADNC), including the intriguing set of individuals with severe/widespread amyloid-β (Aβ) plaques but no/mild tau tangles [Aβ-predominant (AP)-ADNC], may have distinct genetic and clinical features. Analysing National Alzheimer's Coordinating Center data, we stratified 1187 participants into AP-ADNC (n = 95), low Braak primary age-related tauopathy (PART; n = 185), typical-ADNC (n = 832) and high-Braak PART (n = 75). AP-ADNC differed in some clinical features and genetic polymorphisms in the APOE, SNX1, WNT3/MAPT and IGH genes. We conclude that AP-ADNC differs from classical ADNC with implications for in vivo studies.
Collapse
Affiliation(s)
- Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5T 0S8, Canada
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Yuriko Katsumata
- Department of Biostatistics, University of Kentucky, Lexington, KY 40536-0679, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Xian Wu
- Department of Biostatistics, University of Kentucky, Lexington, KY 40536-0679, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Khine Zin Aung
- Department of Biostatistics, University of Kentucky, Lexington, KY 40536-0679, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, Lexington, KY 40536-0679, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Shelley L Forrest
- Tanz Centre for Research in Neurodegenerative Disease and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5T 0S8, Canada
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Pathology, Division of Neuropathology, University of Kentucky, Lexington, KY 40536-0679, USA
| |
Collapse
|
135
|
Coleman PD, Delvaux E, Kordower JH, Boehringer A, Huseby CJ. Massive changes in gene expression and their cause(s) can be a unifying principle in the pathobiology of Alzheimer's disease. Alzheimers Dement 2025; 21:e14555. [PMID: 39912452 PMCID: PMC11851168 DOI: 10.1002/alz.14555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/10/2024] [Accepted: 12/25/2024] [Indexed: 02/07/2025]
Abstract
Understanding of the biology of Alzheimer's disease (AD) has long been fragmented, with various investigators concentrating on amyloid beta (Aβ) or tau, inflammation, cell death pathways, misfolded proteins, glia, and more. Yet data from multiple authors has repeatedly shown altered expression of myriad genes related to these seemingly disparate phenomena. In 2022, Morgan et al. organized the massive data on changes in AD in a meticulous survey of the literature and related these changes to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Their data showed that 91% of the known KEGG pathways are involved in AD and that many of these pathways are represented by the known cellular/molecular phenomena of AD. Such data then raise the fundamental question: What mechanism(s) may be responsible for such widespread changes in gene expression? We review evidence for a unifying model based on sequestrations in stress granules and alteration of nucleocytoplasmic transport in AD. HIGHLIGHTS: In Alzheimer's disease (AD), critical changes take place in neurons before the appearance of plaques or tangles. Addressing these early changes provides a path to early detection and effective intervention in AD.
Collapse
Affiliation(s)
- Paul D. Coleman
- Banner Neurodegenerative Disease Research CenterBiodesign InstituteArizona State UniversityTempeArizonaUSA
| | - Elaine Delvaux
- Banner Neurodegenerative Disease Research CenterBiodesign InstituteArizona State UniversityTempeArizonaUSA
| | - Jeffrey H. Kordower
- Banner Neurodegenerative Disease Research CenterBiodesign InstituteArizona State UniversityTempeArizonaUSA
| | - Ashley Boehringer
- Banner Neurodegenerative Disease Research CenterBiodesign InstituteArizona State UniversityTempeArizonaUSA
| | - Carol J. Huseby
- Banner Neurodegenerative Disease Research CenterBiodesign InstituteArizona State UniversityTempeArizonaUSA
| |
Collapse
|
136
|
Esandi J, Renault P, Capilla-López MD, Blanch R, Edo Á, Ramirez-Gómez D, Bosch A, Almolda B, Saura CA, Giraldo J, Chillón M. HEBE: A novel chimeric chronokine for ameliorating memory deficits in Alzheimer's disease. Biomed Pharmacother 2025; 183:117815. [PMID: 39818099 DOI: 10.1016/j.biopha.2025.117815] [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: 08/09/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/18/2025] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by amyloid-β and Tau protein depositions, with treatments focusing on single proteins have shown limited success due to the complexity of pathways involved. This study explored the potential of chronokines -proteins that modulate aging-related processes- as an alternative therapeutic approach. Specifically, we focused on a novel pleiotropic chimeric protein named HEBE, combining s-KL, sTREM2 and TIMP2, guided by bioinformatic analyses to ensure the preservation of each protein's conformation, crucial for their functions. In vitro studies confirmed HEBE's stability and enzymatic activities, even suggesting it has different activities compared to the individual chronokines. In vivo experiments on APP/Tau mice revealed improved learning and memory functions with HEBE treatment, along with decreased levels of phosphorylated Tau and minor effects on amyloid-β levels. These findings suggest that HEBE is as a promising therapeutic candidate for ameliorating memory deficits and reducing pTau in an AD mouse model.
Collapse
Affiliation(s)
- Jon Esandi
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain.
| | - Pedro Renault
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain; Unitat de Neurociència Translacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bellaterra 08193, Spain.
| | - Maria Dolores Capilla-López
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Rebeca Blanch
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain.
| | - Ángel Edo
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain.
| | - David Ramirez-Gómez
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain.
| | - Assumpció Bosch
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain.
| | - Beatriz Almolda
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain.
| | - Carlos Alberto Saura
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain; Unitat de Neurociència Translacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bellaterra 08193, Spain.
| | - Miguel Chillón
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain.
| |
Collapse
|
137
|
Wu MCS, Wei JHT, Fan RYS, Sim EZ, Yong KT, Gong T, Kong KV. Self-Assembled BODIPY@Au Core-Shell Structures for Durable Neuroprotective Phototherapy. Chembiochem 2025; 26:e202400562. [PMID: 39174489 DOI: 10.1002/cbic.202400562] [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: 06/28/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024]
Abstract
BODIPY analogs are promising photosensitizers for molecular phototherapy; however, they exhibit high dark cytotoxicity and limited singlet oxygen generation capacity. In this study, we developed self-assembled core-shell nanophotosensitizers by linking a bipyridine group to BODIPY (Bpy-BODIPY) and promoting J-aggregation on gold nanourchins. This design enhances photostability and reduces the energy gap between the lowest singlet excited state and the lower triplet state, facilitating efficient singlet oxygen production. We characterized these nanophotosensitizers using UV-visible spectroscopy, transmission electron microscopy (TEM), surface-enhanced Raman spectroscopy (SERS) and dynamic light scattering (DLS), which confirmed the formation of the desired core-shell structure and J-aggregates. Notably, Bpy-BODIPY@Au significantly suppresses tau protein aggregation and enhances neuroprotective action, even in the presence of a phosphatase inhibitor. This work broadens the application of BODIPY chemistry to nanoagents for neuroprotective therapy.
Collapse
Affiliation(s)
- Melody Cai-Syaun Wu
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan R.O.C
| | - Jack Hau-Ting Wei
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan R.O.C
| | - Ricky Yu-Syun Fan
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan R.O.C
| | - Eng Zhi Sim
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan R.O.C
| | - Ken-Tye Yong
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia
| | - Tianxun Gong
- School of Integrated Circuit Science and Engineering (Exemplary School of Microelectronics), University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Kien Voon Kong
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan R.O.C
| |
Collapse
|
138
|
Utpal BK, Roy SC, Zehravi M, Sweilam SH, Raja AD, Haque MA, Nayak C, Balakrishnan S, Singh LP, Panigrahi S, Alshehri MA, Rab SO, Minhaj NS, Emran TB. Polyphenols as Wnt/β-catenin pathway modulators: A promising strategy in clinical neurodegeneration. Animal Model Exp Med 2025; 8:266-286. [PMID: 39808166 PMCID: PMC11871115 DOI: 10.1002/ame2.12525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/18/2024] [Indexed: 01/16/2025] Open
Abstract
Polyphenols, a diverse group of naturally occurring compounds found in plants, have garnered significant attention for their potential therapeutic properties in treating neurodegenerative diseases (NDs). The Wnt/β-catenin (WβC) signaling pathway, a crucial player in neurogenesis, neuronal survival, and synaptic plasticity, is involved in several cellular mechanisms related to NDs. Dysregulation of this pathway is a hallmark in the development of various NDs. This study explores multiple polyphenolic compounds, such as flavonoids, stilbenes, lignans, and phenolic acids, and their potential to protect the nervous system. It provides a comprehensive analysis of their effects on the WβC pathway, elucidating their modes of action. The study highlights the dual function of polyphenols in regulating and protecting the nervous system, providing reassurance about the research benefits. This review provides a comprehensive analysis of the results obtained from both in vitro studies and in vivo research, shedding light on how these substances influence the various components of the pathway. The focus is mainly on the molecular mechanisms that allow polyphenols to reduce oxidative stress, inflammation, and apoptotic processes, ultimately improving the function and survival of neurons. This study aims to offer a thorough understanding of the potential of polyphenols in targeting the WβC signaling pathway, which could lead to the development of innovative therapeutic options for NDs.
Collapse
Affiliation(s)
- Biswajit Kumar Utpal
- Department of Pharmacy, Faculty of Health and Life SciencesDaffodil International UniversityDhakaBangladesh
| | - Sajib Chandra Roy
- Department of Pharmacy, Faculty of PharmacyUniversity of DhakaDhakaBangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry and PharmacyBuraydah Private CollegesBuraydahSaudi Arabia
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of PharmacyPrince Sattam Bin Abdulaziz UniversityAl‐KharjSaudi Arabia
- Department of Pharmacognosy, Faculty of PharmacyEgyptian Russian UniversityCairoEgypt
| | - A. Dinesh Raja
- Department of PharmaceuticsKMCH College of PharmacyCoimbatoreIndia
| | - M. Akiful Haque
- Department of Pharmaceutical Analysis, School of Pharmacy, Anurag University, HyderabadIndia
| | - Chandan Nayak
- Department of Pharmaceutics, School of PharmacyArka Jain UniversityJharkhandIndia
| | - Senthilkumar Balakrishnan
- Department of PharmaceuticsJKKMMRF‐Annai JKK Sampoorani Ammal College of PharmacyKomarapalayamNamakkalIndia
| | - Laliteshwar Pratap Singh
- Department of Pharmaceutical Chemistry, Narayan Institute of PharmacyGopal Narayan Singh UniversitySasaramIndia
| | - Saswati Panigrahi
- Department of Pharmaceutical ChemistrySt. John Institute of Pharmacy and ResearchVevoorPalgharIndia
| | | | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical ScienceKing Khalid UniversityAbhaSaudi Arabia
| | - Najmus Sakib Minhaj
- Department of Pharmacy, Faculty of PharmacyUniversity of DhakaDhakaBangladesh
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life SciencesDaffodil International UniversityDhakaBangladesh
| |
Collapse
|
139
|
Brennan GS, Goriely A. A network aggregation model for amyloid- β dynamics and treatment of Alzheimer's diseases at the brain scale. J Math Biol 2025; 90:22. [PMID: 39891738 PMCID: PMC11787187 DOI: 10.1007/s00285-024-02179-5] [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: 07/01/2024] [Revised: 10/22/2024] [Accepted: 12/22/2024] [Indexed: 02/03/2025]
Abstract
Neurodegenerative diseases are associated with the assembly of specific proteins into oligomers and fibrillar aggregates. At the brain scale, these protein assemblies can diffuse through the brain and seed other regions, creating an autocatalytic protein progression. The growth and transport of these assemblies depend on various mechanisms that can be targeted therapeutically. Here, we use spatially-extended nucleation-aggregation-fragmentation models for the dynamics of prion-like neurodegenerative protein-spreading in the brain to study the effect of different drugs on whole-brain Alzheimer's disease progression.
Collapse
Affiliation(s)
- Georgia S Brennan
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Rd, Oxford, OX2 6GG, UK
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Rd, Oxford, OX2 6GG, UK.
| |
Collapse
|
140
|
Wang X, Zhou L, Liu Y, Ban F, Yang Z, Zhang Y, Hu X, Zhang Y. Screening for Anti-Aβ Aggregation Activity of Marine Fungal Natural Products Based on a Gold Nanoparticle Method. Chem Biodivers 2025; 22:e202401809. [PMID: 39600230 DOI: 10.1002/cbdv.202401809] [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: 07/25/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024]
Abstract
Screening Aβ aggregation inhibitors (AAIs) is important for Alzheimer's disease drug discovery. However, common cellular or biochemical methods are not suitable for high-throughput natural product screening. A gold nanoparticle (GNP) screening method was employed in this study to screen marine fungal crude extracts and pure compounds for quick AAI discovering. The anti-Aβ aggregation activity was further inspected using transmission electron microscopic (TEM) observation, and the interaction between active molecules and different Aβ species was revealed by molecular docking. The results indicated that the fungal extracts DLS2008001(M), BM3T2(M), DLEN2008005(M), TBG1-16(P), and TBG1-13(P) showed activity comparable to the positive control human serum albumin at the concentration of 500 µg/mL; 10 pure compounds also displayed moderate anti-aggregation activity, particularly nidulin, aspergillusidone G, and butyrolactone I. The inspection of anti-Aβ aggregation effect through TEM further demonstrated that extracts TBG1-16(P), DLS2008001(M), and BM3T2(M) dramatically inhibited the formation of Aβ aggregates. Molecular docking displayed low binding energies and key interactions of nidulin, aspergillusidone G, and butyrolactone I, with nine types of Aβ peptides. These findings indicate that the GNP method is efficient in screening AAIs and reveal marine fungal natural products as valuable sources of small molecular AAIs.
Collapse
Affiliation(s)
- Xingyuan Wang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Marine Biomedicine R&D Center at Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Zhanjiang, China
| | - Longjian Zhou
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Marine Biomedicine R&D Center at Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Zhanjiang, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Yayue Liu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Marine Biomedicine R&D Center at Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Zhanjiang, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| | - Fangfang Ban
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Marine Biomedicine R&D Center at Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Zhanjiang, China
| | - Zhiyou Yang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Marine Biomedicine R&D Center at Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Zhanjiang, China
| | - Yongping Zhang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Marine Biomedicine R&D Center at Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Zhanjiang, China
| | - Xueqiong Hu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Marine Biomedicine R&D Center at Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Zhanjiang, China
| | - Yi Zhang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Marine Biomedicine R&D Center at Shenzhen Institute of Guangdong Ocean University, Guangdong Ocean University, Zhanjiang, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
141
|
Schneider LS, Freiesleben SD, van Breukelen G, Wang X, Brosseron F, Heneka MT, Teipel S, Kleineidam L, Stark M, Roy‐Kluth N, Wagner M, Spottke A, Schmid M, Roeske S, Laske C, Munk MH, Perneczky R, Rauchmann B, Buerger K, Janowitz D, Düzel E, Glanz W, Jessen F, Rostamzadeh A, Wiltfang J, Bartels C, Kilimann I, Schneider A, Fliessbach K, Priller J, Spruth EJ, Hellmann‐Regen J, Peters O. Linking higher amyloid beta 1-38 (Aβ(1-38)) levels to reduced Alzheimer's disease progression risk. Alzheimers Dement 2025; 21:e14545. [PMID: 39868793 PMCID: PMC11863357 DOI: 10.1002/alz.14545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025]
Abstract
INTRODUCTION The beneficial effects of amyloid beta 1-38, or Aβ(1-38), on Alzheimer's disease (AD) progression in humans in vivo remain controversial. We investigated AD patients' cerebrospinal fluid (CSF) Aβ(1-38) and AD progression. METHODS Cognitive function and diagnostic change were assessed annually for 3 years in 177 Aβ-positive participants with subjective cognitive decline (SCD), mild cognitive impairment (MCI), and dementia from the German Center for Neurodegenerative Diseases (DZNE) longitudinal cognitive impairment and dementia study (DELCODE) cohort using the Mini-Mental State Examination (MMSE), Preclinical Alzheimer's Cognitive Composite (PACC), Clinical Dementia Rating (CDR), and National Institute of Neurological and Communicative Disorders and Stroke-Alzheimer's Disease and Related Disorders Association (NINCDS-ADRDA) criteria. Mixed linear and Cox regression analyses were conducted. CSF was collected at baseline. RESULTS Higher Aβ(1-38) levels were associated with slower PACC (p = 0.001) and slower CDR Sum of Boxes (CDR-SB) (p = 0.002) but not MMSE decline. Including Aβ(1-40) beyond Aβ(1-38) in the model confirmed an association of Aβ(1-38) with slower PACC decline (p = 0.005), but not with CDR-SB or MMSE decline. In addition, higher Aβ(1-38) baseline levels were associated with a reduced dementia conversion risk. DISCUSSION Further research is needed to understand the role of Aβ(1-38) in AD and its potential for future therapeutic strategies. HIGHLIGHTS This study not only replicates but also extends the existing findings on the role of Aβ(1-38) (amyloid beta 1-38) in Alzheimer's disease (AD) in humans in vivo. Higher baseline Aβ(1-38) levels were associated with a decreased risk of conversion to AD dementia in subjective cognitive decline (SCD) and mild cognitive impairment (MCI). Different linear-mixed regression models suggest an association between higher Aβ(1-38) baseline levels and slower Preclinical Alzheimer's Cognitive Composite (PACC) and Clinical Dementia Rating Sum of Boxes (CDR-SB) decline. Including Aβ(1-40) beyond Aβ(1-38) in the model confirmed a link between Aβ(1-38) and PACC decline, but showed no association of Aβ(1-38) on CDR-SB and Mini-Mental State Examination (MMSE) decline. The impact of short Aβ isoforms in AD progression might have been under-investigated These findings underscore the urgent need for additional research on the role of these shorter Aβ peptides in AD, as they may hold key insights for future therapeutic strategies.
Collapse
|
142
|
Nabizadeh F. Local molecular and connectomic contributions of tau-related neurodegeneration. GeroScience 2025; 47:227-246. [PMID: 39343862 PMCID: PMC11872831 DOI: 10.1007/s11357-024-01339-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
Neurodegeneration in Alzheimer's disease (AD) is known to be mostly driven by tau neurofibrillary tangles. However, both tau and neurodegeneration exhibit variability in their distribution across the brain and among individuals, and the relationship between tau and neurodegeneration might be influenced by several factors. I aimed to map local molecular and connectivity characteristics that affect the association between tau pathology and neurodegeneration. The current study was conducted on the cross-sectional tau-PET and longitudinal T1-weighted MRI scan data of 186 participants from the ADNI dataset including 71 cognitively unimpaired (CU) and 115 mild cognitive impairment (MCI) individuals. Furthermore, the normative molecular profile of a region was defined using neurotransmitter receptor densities, gene expression, T1w/T2w ratio (myelination), FDG-PET (glycolytic index, glucose metabolism, and oxygen metabolism), and synaptic density. I found that the excitatory-inhibitory (E:I) ratio, myelination, synaptic density, glycolytic index, and functional connectivity are linked with deviation in the relationship between tau and neurodegeneration. Furthermore, there was spatial similarity between tau pathology and glycolytic index, synaptic density, and functional connectivity across brain regions. The current study demonstrates that the regional susceptibility to tau-related neurodegeneration is associated with specific molecular and connectomic characteristics of the affected neural systems. I found that the molecular and connectivity architecture of the human brain is linked to the different effects of tau pathology on downstream neurodegeneration.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Alzheimer's Disease Institute, Tehran, Iran.
| |
Collapse
|
143
|
Luo X, Liang J, Lei X, Sun F, Gong M, Liu B, Zhou Z. C/EBPβ in Alzheimer's disease: An integrative regulator of pathological mechanisms. Brain Res Bull 2025; 221:111198. [PMID: 39788461 DOI: 10.1016/j.brainresbull.2025.111198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/22/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Alzheimer's disease (AD) stands as one of the most prevalent neurodegenerative disorders, characterized by a progressive decline in cognitive function, neuroinflammation, amyloid-beta (Aβ) plaques, and neurofibrillary tangles (NFTs). With the global aging population, the incidence of AD continues to rise, yet current therapeutic strategies remain limited in their ability to significantly alleviate cognitive impairments. Therefore, a deeper understanding of the molecular mechanisms underlying AD is imperative for the development of more effective treatments. In recent years, the transcription factor C/EBPβ has emerged as a pivotal regulator in several pathological processes of AD, including neuroinflammation, lipid metabolism, Aβ processing, and tau phosphorylation. Through intricate post-translational modifications, C/EBPβ modulates these processes and may influence the progression of AD on multiple fronts. This review systematically explores the multifaceted roles of C/EBPβ in the pathogenesis of AD, delving into its crucial involvement in neuroinflammation, Aβ production, tau pathology, and lipid metabolism dysregulation. Furthermore, we critically assess therapeutic strategies targeting C/EBPβ, examining the challenges and opportunities in regulating this factor. By synthesizing the latest research findings, we offer a more comprehensive understanding of the role of C/EBPβ in AD and discuss its potential as a therapeutic intervention target.
Collapse
Affiliation(s)
- Xiaoting Luo
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Junyi Liang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Xue Lei
- The First Hospital Affiliated to Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Fengqi Sun
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | | | - Bin Liu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China.
| | - Zhongguang Zhou
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
144
|
Biel D, Suárez-Calvet M, Dewenter A, Steward A, Roemer SN, Dehsarvi A, Zhu Z, Pescoller J, Frontzkowski L, Kreuzer A, Haass C, Schöll M, Brendel M, Franzmeier N. Female sex is linked to a stronger association between sTREM2 and CSF p-tau in Alzheimer's disease. EMBO Mol Med 2025; 17:235-248. [PMID: 39794447 PMCID: PMC11822105 DOI: 10.1038/s44321-024-00190-3] [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: 08/02/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
In Alzheimer's disease (AD), Aβ triggers p-tau secretion, which drives tau aggregation. Therefore, it is critical to characterize modulators of Aβ-related p-tau increases which may alter AD trajectories. Here, we assessed whether factors known to alter tau levels in AD modulate the association between fibrillar Aβ and secreted p-tau181 determined in the cerebrospinal fluid (CSF). To assess potentially modulating effects of female sex, younger age, and ApoE4, we included 322 ADNI participants with cross-sectional/longitudinal p-tau181. To determine effects of microglial activation on p-tau181, we included 454 subjects with cross-sectional CSF sTREM2. Running ANCOVAs for nominal and linear regressions for metric variables, we found that women had higher Aβ-related p-tau181 levels. Higher sTREM2 was associated with elevated p-tau181, with stronger associations in women. Similarly, ApoE4 was related to higher p-tau181 levels and faster p-tau181 increases, with stronger effects in female ApoE4 carriers. Our results show that sex alone modulates the Aβ to p-tau axis, where women show higher Aβ-dependent p-tau secretion, potentially driven by elevated sTREM2-related microglial activation and stronger effects of ApoE4 carriership in women.
Collapse
Affiliation(s)
- Davina Biel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Servei de Neurologia, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Anna Dewenter
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Anna Steward
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Sebastian N Roemer
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Amir Dehsarvi
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Zeyu Zhu
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Julia Pescoller
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Lukas Frontzkowski
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Annika Kreuzer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Schöll
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
| |
Collapse
|
145
|
Elhage A, Cohen S, Cummings J, van der Flier WM, Aisen P, Cho M, Bell J, Hampel H. Defining benefit: Clinically and biologically meaningful outcomes in the next-generation Alzheimer's disease clinical care pathway. Alzheimers Dement 2025; 21:e14425. [PMID: 39697158 PMCID: PMC11848336 DOI: 10.1002/alz.14425] [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: 07/22/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
To understand the potential benefits of emerging Alzheimer's disease (AD) therapies within and beyond clinical trial settings, there is a need to advance current outcome measurements into meaningful information relevant to all stakeholders. The relationship between the impact on disease biology and clinically measurable outcomes in cognition, function, and behavior must be considered when defining the meaningful benefit of early AD therapies. In this review, we discuss: (1) the lack of consideration for biomarkers in the current concept of meaningfulness in AD; (2) the lack of gold standards for determining minimal biologically and clinically important differences (MBCIDs) in AD trials; (3) how the treatment benefits of disease-modifying treatments are cumulative and increase over time; and (4) the different concepts of meaningfulness among key stakeholders. This review utilizes the future clinical biological framework of AD and aims to further integrate and expand the parameters of meaningful benefits toward a precision medicine framework. HIGHLIGHTS: Definition of meaningful benefit from Alzheimer's disease (AD) treatment varies across disease stage and stakeholder perspectives. Observable and meaningful outcomes must consider the clinical-biological nature of AD. Statistically significant effects or outcomes do not always equate to clinically meaningfulness. Assessment tools must reflect stage-specific subtle changes following treatment. Real-world evidence will support consensus, definition, and interpretation of clinical meaningfulness.
Collapse
Affiliation(s)
| | | | | | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
- Amsterdam Neuroscience, NeurodegenerationAmsterdamThe Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Paul Aisen
- Alzheimer's Therapeutic Research Institute, University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Min Cho
- Eisai Inc.NutleyNew JerseyUSA
| | | | | |
Collapse
|
146
|
Krishnamurthy HK, Jayaraman V, Krishna K, Wang T, Bei K, Changalath C, Rajasekaran JJ. An overview of the genes and biomarkers in Alzheimer's disease. Ageing Res Rev 2025; 104:102599. [PMID: 39612989 DOI: 10.1016/j.arr.2024.102599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and neurodegenerative disease characterized by neurofibrillary tangles (NFTs) and amyloid plaque. Familial AD is caused by mutations in the APP, PSEN1, and PSEN2 genes and these mutations result in the early onset of the disease. Sporadic AD usually affects older adults over the age of 65 years and is, therefore classified as late-onset AD (LOAD). Several risk factors associated with LOAD including the APOE gene have been identified. Moreover, GWAS studies have identified a wide array of genes and polymorphisms that are associated with LOAD risk. Currently, the diagnosis of AD involves the evaluation of memory and personality changes, cognitive impairment, and medical and family history to rule out other diseases. Laboratory tests to assess the biomarkers in the body fluids as well as MRI, CT, and PET scans to analyze the presence of plaques and NFTs are also included in the diagnosis of AD. It is important to diagnose AD before the onset of clinical symptoms, i.e. during the preclinical stage, to delay the progression and for better management of the disease. Research has been conducted to identify biomarkers of AD in the CSF, serum, saliva, and urine during the preclinical stage. Current research has identified several biomarkers and potential biomarkers in the body fluids that enhance diagnostic accuracy. Aside from genetics, other factors such as diet, physical activity, and lifestyle factors may influence the risk of developing AD. Clinical trials are underway to find potential biomarkers, diagnostic measures, and treatments for AD mainly in the preclinical stage. This review provides an overview of the genes and biomarkers of AD.
Collapse
Affiliation(s)
| | | | - Karthik Krishna
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | - Tianhao Wang
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | - Kang Bei
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | | | | |
Collapse
|
147
|
Hao M, Chu J, Zhang T, Yin T, Gu Y, Liang W, Ji W, Zhuang J, Liu Y, Gao J, Yin Y. Nanomaterials-mediated lysosomal regulation: a robust protein-clearance approach for the treatment of Alzheimer's disease. Neural Regen Res 2025; 20:424-439. [PMID: 38819046 PMCID: PMC11317947 DOI: 10.4103/nrr.nrr-d-23-01736] [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: 10/22/2023] [Revised: 01/04/2024] [Accepted: 02/20/2024] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is a debilitating, progressive neurodegenerative disorder characterized by the progressive accumulation of abnormal proteins, including amyloid plaques and intracellular tau tangles, primarily within the brain. Lysosomes, crucial intracellular organelles responsible for protein degradation, play a key role in maintaining cellular homeostasis. Some studies have suggested a link between the dysregulation of the lysosomal system and pathogenesis of neurodegenerative diseases, including Alzheimer's disease. Restoring the normal physiological function of lysosomes hold the potential to reduce the pathological burden and improve the symptoms of Alzheimer's disease. Currently, the efficacy of drugs in treating Alzheimer's disease is limited, with major challenges in drug delivery efficiency and targeting. Recently, nanomaterials have gained widespread use in Alzheimer's disease drug research owing to their favorable physical and chemical properties. This review aims to provide a comprehensive overview of recent advances in using nanomaterials (polymeric nanomaterials, nanoemulsions, and carbon-based nanomaterials) to enhance lysosomal function in treating Alzheimer's disease. This review also explores new concepts and potential therapeutic strategies for Alzheimer's disease through the integration of nanomaterials and modulation of lysosomal function. In conclusion, this review emphasizes the potential of nanomaterials in modulating lysosomal function to improve the pathological features of Alzheimer's disease. The application of nanotechnology to the development of Alzheimer's disease drugs brings new ideas and approaches for future treatment of this disease.
Collapse
Affiliation(s)
- Mengqi Hao
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jianjian Chu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tong Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Yuankai Gu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Wendanqi Liang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wenbo Ji
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
148
|
Kepp KP, Høilund-Carlsen PF, Cristea IA, Cumming RG, Daly T, Emilsson L, Flacco ME, Hemkens LG, Janiaud P, Johnsen KB, Lallukka T, Muka T, Naudet F, Revheim MER, Squitti R, Thambisetty M. Communicating scientific evidence: drugs for Alzheimer's disease as a case study. Curr Med Res Opin 2025; 41:347-354. [PMID: 39853750 DOI: 10.1080/03007995.2025.2458530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/19/2024] [Accepted: 01/21/2025] [Indexed: 01/26/2025]
Abstract
This paper reviews the scientific evidence on new anti-amyloid monoclonal antibodies for treating Alzheimer's disease as a case study for improving scientific evidence communication. We introduce five guidelines condensed from the biomedical evidence literature but adapted to the short format of science communication in e.g. journal opinion pieces and newspaper articles. Given the major importance and recent confusion regarding the discussed drugs, with certain disagreements seen e.g. between FDA and EMA, the suggested guidelines may be useful to clinicians discussing with their patients and to scientists communicating the evidence in balance. More generally, we hope that the guidelines may help us to improve communication of scientific evidence on complex topics in opinion pieces in the scientific literature, in advocacy, and in media appearances.
Collapse
Affiliation(s)
| | - Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ioana A Cristea
- Department of General Psychology, University of Padova, Padova, Italy
| | - Robert G Cumming
- School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Timothy Daly
- Bioethics Program, FLACSO Argentina, Buenos Aires, Argentina
- UMR 1219, Bordeaux Population Health, University of Bordeaux & INSERM, Bordeaux, France
| | - Louise Emilsson
- General Practice Research Unit (AFE) and Department of General Practice, Institute of Health and Society, University of Oslo, Oslo, Norway
- Vårdcentralen Värmlands Nysäter and Centre for Clinical Research, County Council of Värmland, Värmland, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Solna, Sweden
| | - Maria E Flacco
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| | - Lars G Hemkens
- Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Perrine Janiaud
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | | | - Tea Lallukka
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | | | - Florian Naudet
- Institut de recherche en santé, environnement et travail, Inserm, EHESP, Centre d'investigation clinique de Rennes (CIC1414), University of Rennes, Rennes, France
- Institut Universitaire de France, Paris, France
| | - Mona-Elisabeth R Revheim
- The Intervention Centre, Division of Technology and Innovation, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Rosanna Squitti
- Department of Laboratory Science, Research and Development Division, Ospedale Isola Tiberina Gemelli Isola, Rome, Italy
- Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Como, Italy
| | - Madhav Thambisetty
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
149
|
Wang W, Myers SJ, Ollen-Bittle N, Whitehead SN. Elevation of ganglioside degradation pathway drives GM2 and GM3 within amyloid plaques in a transgenic mouse model of Alzheimer's disease. Neurobiol Dis 2025; 205:106798. [PMID: 39793768 DOI: 10.1016/j.nbd.2025.106798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/05/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that accounts for two-thirds of all dementia cases, and age is the strongest risk factor. In addition to the amyloid hypothesis, lipid dysregulation is now recognized as a core component of AD pathology. Gangliosides are a class of membrane lipids of the glycosphingolipid family and are enriched in the central nervous system (CNS). Ganglioside dysregulation has been implicated in various neurodegenerative diseases, including AD, but the spatial distribution of ganglioside dysregulation with respect to amyloid-beta (Aβ) deposition is not well understood. To address this gap, matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) was employed to investigate the age-dependent expression profiles of the A-series ganglioside species GD1a, GM1, GM2, and GM3 in the APP/PS1 transgenic mouse model of AD in which age-dependent amyloid-beta (Aβ) plaques develop. This study utilized a dual-resolution approach in combination with whole-brain imaging for comprehensive detection of ganglioside expression across neuroanatomical regions via high-resolution imaging of the cerebral cortex and hippocampus to investigate plaque-associated ganglioside alterations. The results revealed age-dependent changes in the complex gangliosides GM1 and GD1a across white and gray matter regions in both wildtype and APP/PS1 mice. Significantly greater levels of simple gangliosides GM2 and GM3 were observed in the cortex and dentate gyrus of the hippocampus in transgenic mice at 12 and 18 m than in age-matched controls. The accumulation of GM3 colocalized with Aβ plaques in aged APP/PS1 mice and correlated with Hexa gene expression, suggesting that ganglioside degradation is a mechanism for the accumulation of GM3. This work is the first to demonstrate that age-related ganglioside dysregulation is spatiotemporally associated with Aβ plaques using sophisticated MSI and reveals novel mechanistic insights into lipid regulation in AD.
Collapse
Affiliation(s)
- Wenxuan Wang
- Vulnerable Brain Lab, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Sarah J Myers
- Vulnerable Brain Lab, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Nikita Ollen-Bittle
- Vulnerable Brain Lab, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Shawn N Whitehead
- Vulnerable Brain Lab, Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada.
| |
Collapse
|
150
|
Yu Z, Liu J, Liu Z, Liu X, Tuo J, Li J, Tu Y, Tan Q, Ma Y, Bai Y, Xin J, Huang S, Zeng G, Shi A, Wang J, Liu Y, Bu X, Ye L, Wan Y, Liu T, Chen X, Qiu Z, Gao C, Wang Y. Roles of blood monocytes carrying TREM2 R47H mutation in pathogenesis of Alzheimer's disease and its therapeutic potential in APP/PS1 mice. Alzheimers Dement 2025; 21:e14402. [PMID: 39740209 PMCID: PMC11848385 DOI: 10.1002/alz.14402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 09/27/2024] [Accepted: 10/21/2024] [Indexed: 01/02/2025]
Abstract
INTRODUCTION The triggering receptor expressed on myeloid cells 2 (TREM2) arginine-47-histidine (R47H) mutation is a significant risk for Alzheimer's disease (AD) with unclear mechanisms. Previous studies focused on microglial amyloid-β (Aβ) phagocytosis with less attention on the impact of TREM2R47H mutation on blood monocytes. METHODS Bone marrow transplantation (BMT) models were used to assess the contribution of blood monocytes carrying TREM2R47H mutation to AD. RESULTS Aβ phagocytosis was compromised in mouse monocytes carrying the TREM2R47H mutation. Transplantation of bone marrow cells (BMCs) carrying TREM2R47H mutation increased cerebral Aβ burden and aggravated AD-type pathologies. Moreover, the replacement of TREM2R47H-BMCs restored monocytic Aβ phagocytosis, lowered Aβ levels in the blood and brain, and improved cognitive function. DISCUSSION Our study reveals that blood monocytes carrying the TREM2R47H mutation substantially contribute to the pathogenesis of AD, and correcting the TREM2R47H mutation in BMCs would be a potential therapeutic approach for those carrying this mutation. HIGHLIGHTS TREM2R47H mutation compromises the Aβ phagocytosis of blood monocytes. Blood monocytes carrying TREM2R47H mutation contribute substantially to AD pathogenesis. Correction of the TREM2R47H mutation in bone marrow cells ameliorates AD pathologies and cognitive impairments.
Collapse
Affiliation(s)
- Zhong‐Yuan Yu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Institute of Brain and IntelligenceChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Jie Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Institute of Brain and IntelligenceChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Zhi‐Hao Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiao‐Yu Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Department of NeurologyThe 991st Hospital of Chinese People's Liberation Army Joint Logistic Support ForceXiangyangChina
| | - Jin‐Mei Tuo
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Jiang‐Hui Li
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Yun‐Feng Tu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Qi Tan
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Yuan‐Yuan Ma
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Yu‐Di Bai
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Jia‐Yan Xin
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Shan Huang
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Gui‐Hua Zeng
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - An‐Yu Shi
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Jun Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Yu‐Hui Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Xian‐Le Bu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Li‐Lin Ye
- Institute of ImmunologyThird Military Medical UniversityChongqingChina
| | - Ying Wan
- Biomedical Analysis CentreThird Military Medical UniversityChongqingChina
| | - Tong‐Fei Liu
- Institute for Brain Science and DiseaseChongqing Medical UniversityChongqingChina
| | - Xiao‐Wei Chen
- Institute of Brain and IntelligenceChongqingChina
- Brain Research CentreCollaborative Innovation Centre for Brain ScienceThird Military Medical UniversityChongqingChina
| | - Zi‐Long Qiu
- Songjiang HospitalSongjiang InstituteShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chang‐Yue Gao
- Department of Rehabilitation MedicineDaping Hospital, Third Military Medical UniversityChongqingChina
| | - Yan‐Jiang Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, Third Military Medical UniversityChongqingChina
- Institute of Brain and IntelligenceChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|