1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [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: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Fu H, Li J, Zhang C, Gao G, Ge Q, Guan X, Cui D. Pathological axonal enlargement in connection with amyloidosis, lysosome destabilization, and bleeding is a major defect in Alzheimer's disease. Neural Regen Res 2026; 21:790-799. [PMID: 40326989 DOI: 10.4103/nrr.nrr-d-24-01440] [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: 11/18/2024] [Accepted: 03/17/2025] [Indexed: 05/07/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00047/figure1/v/2025-05-05T160104Z/r/image-tiff Alzheimer's disease is a multi-amyloidosis disease characterized by amyloid-β deposits in brain blood vessels, microaneurysms, and senile plaques. How amyloid-β deposition affects axon pathology has not been examined extensively. We used immunohistochemistry and immunofluorescence staining to analyze the forebrain tissue slices of Alzheimer's disease patients. Widespread axonal amyloidosis with distinctive axonal enlargement was observed in patients with Alzheimer's disease. On average, amyloid-β-positive axon diameters in Alzheimer's disease brains were 1.72 times those of control brain axons. Furthermore, axonal amyloidosis was associated with microtubule-associated protein 2 reduction, tau phosphorylation, lysosome destabilization, and several blood-related markers, such as apolipoprotein E, alpha-hemoglobin, glycosylated hemoglobin type A1C, and hemin. Lysosome destabilization in Alzheimer's disease was also clearly identified in the neuronal soma, where it was associated with the co-expression of amyloid-β, Cathepsin D, alpha-hemoglobin, actin alpha 2, and collagen type IV. This suggests that exogenous hemorrhagic protein intake influences neural lysosome stability. Additionally, the data showed that amyloid-β-containing lysosomes were 2.23 times larger than control lysosomes. Furthermore, under rare conditions, axonal breakages were observed, which likely resulted in Wallerian degeneration. In summary, axonal enlargement associated with amyloidosis, micro-bleeding, and lysosome destabilization is a major defect in patients with Alzheimer's disease. This finding suggests that, in addition to the well-documented neural soma and synaptic damage, axonal damage is a key component of neuronal defects in Alzheimer's disease.
Collapse
Affiliation(s)
- Hualin Fu
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute of Marine Equipment, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jilong Li
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chunlei Zhang
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guo Gao
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiqi Ge
- Institute of Marine Equipment, Shanghai Jiao Tong University, Shanghai, China
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China
| | - Xinping Guan
- Department of Automation, Shanghai Jiao Tong University, Shanghai, China
- The Key Laboratory of System Control and Information Processing, Ministry of Education, Shanghai, China
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
3
|
Sha S, Ren L, Xing X, Guo W, Wang Y, Li Y, Cao Y, Qu L. Recent advances in immunotherapy targeting amyloid-beta and tauopathies in Alzheimer's disease. Neural Regen Res 2026; 21:577-587. [PMID: 39885674 DOI: 10.4103/nrr.nrr-d-24-00846] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/28/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease, a devastating neurodegenerative disorder, is characterized by progressive cognitive decline, primarily due to amyloid-beta protein deposition and tau protein phosphorylation. Effectively reducing the cytotoxicity of amyloid-beta42 aggregates and tau oligomers may help slow the progression of Alzheimer's disease. Conventional drugs, such as donepezil, can only alleviate symptoms and are not able to prevent the underlying pathological processes or cognitive decline. Currently, active and passive immunotherapies targeting amyloid-beta and tau have shown some efficacy in mice with asymptomatic Alzheimer's disease and other transgenic animal models, attracting considerable attention. However, the clinical application of these immunotherapies demonstrated only limited efficacy before the discovery of lecanemab and donanemab. This review first discusses the advancements in the pathogenesis of Alzheimer's disease and active and passive immunotherapies targeting amyloid-beta and tau proteins. Furthermore, it reviews the advantages and disadvantages of various immunotherapies and considers their future prospects. Although some antibodies have shown promise in patients with mild Alzheimer's disease, substantial clinical data are still lacking to validate their effectiveness in individuals with moderate Alzheimer's disease.
Collapse
Affiliation(s)
- Sha Sha
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Lina Ren
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaona Xing
- Department of Neurology, Shenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China
| | - Wanshu Guo
- Department of Neurology, People's Hospital of Liaoning Province, Shenyang, Liaoning Province, China
| | - Yan Wang
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ying Li
- Department of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yunpeng Cao
- Department of Neurology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Le Qu
- Department of Dermatology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
4
|
Sun Y, Pang X, Huang X, Liu D, Huang J, Zheng P, Wei Y, Pang C. Potential mechanisms of non-coding RNA regulation in Alzheimer's disease. Neural Regen Res 2026; 21:265-280. [PMID: 39851253 PMCID: PMC12094571 DOI: 10.4103/nrr.nrr-d-24-00696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/02/2024] [Accepted: 10/19/2024] [Indexed: 01/26/2025] Open
Abstract
Alzheimer's disease, a progressively degenerative neurological disorder, is the most common cause of dementia in the elderly. While its precise etiology remains unclear, researchers have identified diverse pathological characteristics and molecular pathways associated with its progression. Advances in scientific research have increasingly highlighted the crucial role of non-coding RNAs in the progression of Alzheimer's disease. These non-coding RNAs regulate several biological processes critical to the advancement of the disease, offering promising potential as therapeutic targets and diagnostic biomarkers. Therefore, this review aims to investigate the underlying mechanisms of Alzheimer's disease onset, with a particular focus on microRNAs, long non-coding RNAs, and circular RNAs associated with the disease. The review elucidates the potential pathogenic processes of Alzheimer's disease and provides a detailed description of the synthesis mechanisms of the three aforementioned non-coding RNAs. It comprehensively summarizes the various non-coding RNAs that have been identified to play key regulatory roles in Alzheimer's disease, as well as how these non-coding RNAs influence the disease's progression by regulating gene expression and protein functions. For example, miR-9 targets the UBE4B gene, promoting autophagy-mediated degradation of Tau protein, thereby reducing Tau accumulation and delaying Alzheimer's disease progression. Conversely, the long non-coding RNA BACE1-AS stabilizes BACE1 mRNA, promoting the generation of amyloid-β and accelerating Alzheimer's disease development. Additionally, circular RNAs play significant roles in regulating neuroinflammatory responses. By integrating insights from these regulatory mechanisms, there is potential to discover new therapeutic targets and potential biomarkers for early detection and management of Alzheimer's disease. This review aims to enhance the understanding of the relationship between Alzheimer's disease and non-coding RNAs, potentially paving the way for early detection and novel treatment strategies.
Collapse
Affiliation(s)
- Yue Sun
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Xinping Pang
- School of Science, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu Province, China
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Dinglu Liu
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Jingyue Huang
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Pengtao Zheng
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| | - Yanyu Wei
- National Key Laboratory of Science and Technology on Vacuum Electronics, School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Chaoyang Pang
- College of Computer Science, Sichuan Normal University, Chengdu, Sichuan Province, China
| |
Collapse
|
5
|
Riffo-Lepe N, González-Sanmiguel J, Armijo-Weingart L, Saavedra-Sieyes P, Hernandez D, Ramos G, San Martín LS, Aguayo LG. Synaptic and synchronic impairments in subcortical brain regions associated with early non-cognitive dysfunction in Alzheimer's disease. Neural Regen Res 2026; 21:248-264. [PMID: 39885666 PMCID: PMC12094569 DOI: 10.4103/nrr.nrr-d-24-01052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/16/2024] [Accepted: 12/21/2024] [Indexed: 02/01/2025] Open
Abstract
For many decades, Alzheimer's disease research has primarily focused on impairments within cortical and hippocampal regions, which are thought to be related to cognitive dysfunctions such as memory and language deficits. The exact cause of Alzheimer's disease is still under debate, making it challenging to establish an effective therapy or early diagnosis. It is widely accepted that the accumulation of amyloid-beta peptide in the brain parenchyma leads to synaptic dysfunction, a critical step in Alzheimer's disease development. The traditional amyloid cascade model is initiated by accumulating extracellular amyloid-beta in brain areas essential for memory and language. However, while it is possible to reduce the presence of amyloid-beta plaques in the brain with newer immunotherapies, cognitive symptoms do not necessarily improve. Interestingly, recent studies support the notion that early alterations in subcortical brain regions also contribute to brain damage and precognitive decline in Alzheimer's disease. A body of recent evidence suggests that early Alzheimer's disease is associated with alterations (e.g., motivation, anxiety, and motor impairment) in subcortical areas, such as the striatum and amygdala, in both human and animal models. Also, recent data indicate that intracellular amyloid-beta appears early in subcortical regions such as the nucleus accumbens, locus coeruleus, and raphe nucleus, even without extracellular amyloid plaques. The reported effects are mainly excitatory, increasing glutamatergic transmission and neuronal excitability. In agreement, data in Alzheimer's disease patients and animal models show an increase in neuronal synchronization that leads to electroencephalogram disturbances and epilepsy. The data indicate that early subcortical brain dysfunctions might be associated with non-cognitive symptoms such as anxiety, irritability, and motivation deficits, which precede memory loss and language alterations. Overall, the evidence reviewed suggests that subcortical brain regions could explain early dysfunctions and perhaps be targets for therapies to slow disease progression. Future research should focus on these non-traditional brain regions to reveal early pathological alterations and underlying mechanisms to advance our understanding of Alzheimer's disease beyond the traditionally studied hippocampal and cortical circuits.
Collapse
Affiliation(s)
- Nicolás Riffo-Lepe
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Juliana González-Sanmiguel
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Lorena Armijo-Weingart
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Concepción, Chile
| | - Paulina Saavedra-Sieyes
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - David Hernandez
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Gerson Ramos
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Loreto S. San Martín
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| | - Luis G. Aguayo
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| |
Collapse
|
6
|
Xun QQ, Zhang J, Feng L, Ma YY, Li Y, Shi XL. Identification of a novel pyrrolo[2,3- b]pyridine compound as a potent glycogen synthase kinase 3β inhibitor for treating Alzheimer's disease. J Enzyme Inhib Med Chem 2025; 40:2466846. [PMID: 39976249 PMCID: PMC11843656 DOI: 10.1080/14756366.2025.2466846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
Herein, a novel pyrrolo[2,3-b]pyridine-based glycogen synthase kinase 3β (GSK-3β) inhibitor, S01, was rationally designed and synthesised to target Alzheimer's disease (AD). S01 inhibited GSK-3β, with an IC50 of 0.35 ± 0.06 nM, and had an acceptable kinase selectivity for 24 structurally similar kinases. Western blotting assays indicated that S01 efficiently increased the expression of p-GSK-3β-Ser9 and decreased p-tau-Ser396 levels in a dose-dependent manner. In vitro cell experiments, S01 showed low cytotoxicity to SH-SY5Y cells, significantly upregulated the expression of β-catenin and neurogenesis-related biomarkers, and effectively promoted the outgrowth of differentiated neuronal neurites. Moreover, S01 substantially ameliorated dyskinesia in AlCl3-induced zebrafish AD models at a concentration of 0.12 μM, which was more potent than Donepezil (8 μM) under identical conditions. Acute toxicity experiments further confirmed the safety of S01 in vivo. Our findings suggested that S01 is a prospective GSK-3β inhibitor and can be tested as a candidate for treating AD.
Collapse
Affiliation(s)
- Qing-Qing Xun
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
- School of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Jing Zhang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Lei Feng
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yu-Ying Ma
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Ying Li
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Xiao-Long Shi
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
7
|
Jin J, Zhang H, Lu Q, Tian L, Yao S, Lai F, Liang Y, Liu C, Lu Y, Tian S, Zhao Y, Ren W. Nanocarrier-mediated siRNA delivery: a new approach for the treatment of traumatic brain injury-related Alzheimer's disease. Neural Regen Res 2025; 20:2538-2555. [PMID: 39314170 PMCID: PMC11801294 DOI: 10.4103/nrr.nrr-d-24-00303] [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: 03/16/2024] [Revised: 07/06/2024] [Accepted: 07/17/2024] [Indexed: 09/25/2024] Open
Abstract
Traumatic brain injury and Alzheimer's disease share pathological similarities, including neuronal loss, amyloid-β deposition, tau hyperphosphorylation, blood-brain barrier dysfunction, neuroinflammation, and cognitive deficits. Furthermore, traumatic brain injury can exacerbate Alzheimer's disease-like pathologies, potentially leading to the development of Alzheimer's disease. Nanocarriers offer a potential solution by facilitating the delivery of small interfering RNAs across the blood-brain barrier for the targeted silencing of key pathological genes implicated in traumatic brain injury and Alzheimer's disease. Unlike traditional approaches to neuroregeneration, this is a molecular-targeted strategy, thus avoiding non-specific drug actions. This review focuses on the use of nanocarrier systems for the efficient and precise delivery of siRNAs, discussing the advantages, challenges, and future directions. In principle, siRNAs have the potential to target all genes and non-targetable proteins, holding significant promise for treating various diseases. Among the various therapeutic approaches currently available for neurological diseases, siRNA gene silencing can precisely "turn off" the expression of any gene at the genetic level, thus radically inhibiting disease progression; however, a significant challenge lies in delivering siRNAs across the blood-brain barrier. Nanoparticles have received increasing attention as an innovative drug delivery tool for the treatment of brain diseases. They are considered a potential therapeutic strategy with the advantages of being able to cross the blood-brain barrier, targeted drug delivery, enhanced drug stability, and multifunctional therapy. The use of nanoparticles to deliver specific modified siRNAs to the injured brain is gradually being recognized as a feasible and effective approach. Although this strategy is still in the preclinical exploration stage, it is expected to achieve clinical translation in the future, creating a new field of molecular targeted therapy and precision medicine for the treatment of Alzheimer's disease associated with traumatic brain injury.
Collapse
Affiliation(s)
- Jie Jin
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Huajing Zhang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Linqiang Tian
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
- Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province, China
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Feng Lai
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Yangfan Liang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Chuanchuan Liu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yujia Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Sijia Tian
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Wenjie Ren
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
- Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province, China
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province, China
| |
Collapse
|
8
|
Higashi Y, Ikuno K, Saito T, Saido TC, Miyasaka T, Kakuda N, Funamoto S. High Affinity Staining for Histological Immunoreactivity revealed phosphorylated tau within amyloid-cored plaques in the brain of AD model mice. Biochem Biophys Res Commun 2025; 771:152025. [PMID: 40393159 DOI: 10.1016/j.bbrc.2025.152025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025]
Abstract
The historical pathology of the brain in Alzheimer's disease (AD) is characterized by the amyloid cascade hypothesis, in which amyloid β protein accumulates in the extracellular parenchyma as senile plaque and triggers phosphorylation of microtubule-associated protein tau for forming neurofibrillary tangle in the human neurons. Whether these protein existences differed in the brain parenchyma, the relationship of these proteins of accumulation mechanisms is unknown. In the case of brain pathological analysis, the level of phosphorylation for tau has been decreased in the paraffin-embedded sections compared with biochemical analysis. Here, we have established and developed a method to highlight phosphorylated proteins including tau with frozen sections, as the HIGh Affinity Staining Histological Immunoreactivity (HIGASHI) method. Using this HIGASHI method, hyper-phosphorylated tau could be detected in the mossy fiber on the frozen brain sections of wild-type mice under hypothermia conditions. Here, we attempted the HIGASHI method to detect senile plaque and phosphorylated tau in the AD model mouse brains. Phosphorylated tau was found in the center of senile plaques in the mice brain parenchyma. Additionally, these senile plaques colocalized with microglia cells in the center of senile plaques. Interestingly, senile plaques have been made in the tau knock-out mice brains expressing human amyloid precursor protein. Thus, senile plaques have been composed of Aβ and phosphorylated tau in the brain, but tau isn't necessary for bearing senile plaques.
Collapse
Affiliation(s)
- Yuto Higashi
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan; Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan
| | - Kanta Ikuno
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan; Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8602, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Tomohiro Miyasaka
- Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan; Laboratory of Physiology and Anatomy, Nihon University, School of Pharmacy, Funabashi, Chiba, 274-8555, Japan.
| | - Nobuto Kakuda
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan; Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan.
| | - Satoru Funamoto
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan; Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan.
| |
Collapse
|
9
|
Salgado KDCB, Nascimento RGF, Albuquerque ALS, Oliveira LAM, Pinto Coelho Nogueira KDO. Melatonin protects mouse hippocampal neurons from neurotoxicity induced by amyloid β-peptide 25-35. Brain Res 2025; 1859:149637. [PMID: 40222698 DOI: 10.1016/j.brainres.2025.149637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder and the leading cause of dementia in the elderly, as classified by the WHO. Its neuropathological hallmarks include the accumulation of amyloid-β (Aβ) plaques and intracellular tau tangles, which contribute to oxidative stress, mitochondrial dysfunction, lipid peroxidation, and neuronal death. Emerging evidence suggests that melatonin, a potent antioxidant produced by the pineal gland, plays a neuroprotective role in AD, yet its precise mechanisms remain underexplored. In this study, we utilized a physiologically relevant primary culture of hippocampal neurons to investigate melatonin's protective effects against toxicity induced by Aβ25-35. Our findings demonstrate that melatonin significantly enhances cellular metabolism and viability while reducing reactive oxygen species (ROS) levels and lipid peroxidation, thereby mitigating Aβ-induced neurotoxicity. These results provide mechanistic insights into melatonin's antioxidative and neuroprotective properties, reinforcing its potential as a therapeutic agent against oxidative stress in AD. This study underscores the promise of melatonin-based interventions in the development of novel antioxidant-targeted therapies for AD.
Collapse
Affiliation(s)
- Karen Del Carmen B Salgado
- Department of Biological Sciences, Laboratory of Neurobiology and Biomaterials (LNBio), Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Rosiene G F Nascimento
- Department of Biological Sciences, Laboratory of Neurobiology and Biomaterials (LNBio), Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Ana Luiza S Albuquerque
- Department of Biological Sciences, Laboratory of Neurobiology and Biomaterials (LNBio), Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Laser A M Oliveira
- Department of Biological Sciences, Laboratory of Neurobiology and Biomaterials (LNBio), Federal University of Ouro Preto, Ouro Preto, Brazil
| | | |
Collapse
|
10
|
Najdek C, Walle P, Flaig A, Ayral AM, Demiautte F, Coulon A, Buiche V, Lambert E, Amouyel P, Gelle C, Siedlecki-Wullich D, Dumont J, Kilinc D, Eysert F, Lambert JC, Chapuis J. Calpain and caspase regulate Aβ peptide production via cleavage of KINDLIN2 encoded by the AD-associated gene FERMT2. Neurobiol Aging 2025; 151:117-125. [PMID: 40273529 DOI: 10.1016/j.neurobiolaging.2025.04.009] [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: 04/14/2025] [Accepted: 04/19/2025] [Indexed: 04/26/2025]
Abstract
The adapter protein KINDLIN2, encoded by the Alzheimer's disease (AD) genetic risk factor FERMT2, was identified as a modulator of APP processing. KINDLIN2 directly interacts with APP to modulate its metabolism, and KINDLIN2 underexpression impairs long-term potentiation in an APP-dependent manner. Altogether, these data suggest that loss of KINDLIN2 could have a detrimental effect on synaptic function and promote AD pathophysiological process. In this study, we identified KINDLIN2 as a novel substrate of caspases and calpain I, two well-characterized cysteine proteases involved in the regulation of synaptic plasticity. These cleavages resulted in the dissociation of the F0 and F1 domains of KINDLIN2 that are necessary for it to function as an adapter protein. Furthermore, we demonstrate that these cleavages lead to a decrease in KINDLIN2's ability to control APP processing. Overall, these KINDLIN2 cleavages appear as potential new mechanisms in the regulation of KINDLIN2 functions at the synapse and could be of interest for the pathophysiology of AD.
Collapse
Affiliation(s)
- Chloé Najdek
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Pauline Walle
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Amandine Flaig
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Anne-Marie Ayral
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Florie Demiautte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Audrey Coulon
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Valérie Buiche
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Erwan Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Carla Gelle
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Dolores Siedlecki-Wullich
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Julie Dumont
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Devrim Kilinc
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Fanny Eysert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France
| | - Julien Chapuis
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille 59019, France.
| |
Collapse
|
11
|
Chen J, Wang X, Liu X, Shi L, Yu XQ, Cen X, Li K. Rational Design of Long-Circulating Bright Fluorescent Probe for In Vivo Imaging of Amyloid-β Plaques in Alzheimer's Disease. Anal Chem 2025; 97:12347-12355. [PMID: 40377489 DOI: 10.1021/acs.analchem.5c01619] [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: 05/18/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the pathological accumulation of amyloid-β (Aβ) plaques, which serve as crucial biomarkers for disease diagnosis and therapeutic evaluation. While fluorescence imaging has emerged as a powerful technique for Aβ detection, current probes face limitations in clinical application due to insufficient photostability and short blood half-life, resulting in compromised signal-to-noise ratios (SNRs) and imaging resolution. Herein, two bright quinoxalinone-based fluorescent probes (QNO-AD-PEGs) were presented, which incorporate hydrophilic poly(ethylene glycol) (PEG) chains for enhanced biocompatibility and an Aβ-specific N,N-dimethylaminophenyl recognition unit. QNO-AD-PEG1 demonstrated exceptional binding affinity for Aβ42 aggregates (Kd = 42 nM) and a remarkable 49-fold fluorescence enhancement upon target engagement, with a quantum yield (ΦAβ) of 11.45%. In vivo imaging revealed that QNO-AD-PEG1 effectively crossed the blood-brain barrier (BBB) and exhibited a prolonged half-life (315 min). Notably, the probe successfully visualized age-dependent Aβ plaque progression in AD mouse models. This study presents a significant breakthrough in molecular imaging for neurodegenerative diseases, offering a versatile tool for both fundamental AD research and potential clinical applications.
Collapse
Affiliation(s)
- Jie Chen
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, P. R. China
| | - Xiaojie Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- Department of Neuroscience, City University of HongKong, Hong Kong 523808, P. R. China
| | - Xinyao Liu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, P. R. China
| | - Lei Shi
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, P. R. China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China
| | - Xiao-Qi Yu
- Department of Chemistry, Xihua University, Chengdu 610039, P. R. China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Medical School, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, P. R. China
| |
Collapse
|
12
|
Albrahadi T, Hureau C, Platts JA. Constant pH Molecular Dynamics Simulation of pH Effects on Amyloid-β Structure, Dynamics, and Metal-Binding. Chemistry 2025; 31:e202500547. [PMID: 40331332 DOI: 10.1002/chem.202500547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 05/01/2025] [Accepted: 05/05/2025] [Indexed: 05/08/2025]
Abstract
We report the first molecular dynamics simulations to examine the effect of pH on the structure, dynamics, and metal-binding ability of amyloid-β, the peptide implicated in the onset of Alzheimer's disease. We show that in the pH range of 6 to 8 only histidine residues show variable protonation, that predicted pKa values are in agreement with experimental data, and that changes in pH affect the size, flexibility, and secondary structure of the peptide. The binding of Cu(II) or Zn(II) to the peptide induces a shift of 1 to 1.5 pKa units in unbound histidine residues, while metal binding modes associated with higher pH induce significant changes in peptide structure. We speculate on the significance of these findings on results showing pH dependence as well as on Cu(II) and Zn(II) modulation of aggregation of Amyloid-β.
Collapse
Affiliation(s)
- Thuraya Albrahadi
- School of Chemistry, Cardiff University, Park Place, Cardiff, CF10 3AT, UK
| | - Christelle Hureau
- Laboratoire de Chimie de Coordination - CNRS UPR8241, Université de Toulouse, Toulouse, 31000, France
| | - James A Platts
- School of Chemistry, Cardiff University, Park Place, Cardiff, CF10 3AT, UK
| |
Collapse
|
13
|
Pedrinolla A, Dorelli G, Porcelli S, Burleigh M, Mendo M, Martignon C, Fonte C, Dalle Carbonare LG, Easton C, Muti E, Schena F, Venturelli M. Increasing nitric oxide availability via ingestion of nitrate-rich beetroot juice improves vascular responsiveness in individuals with Alzheimer's Disease. Nitric Oxide 2025; 156:50-56. [PMID: 40089052 DOI: 10.1016/j.niox.2025.03.001] [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: 10/21/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Poor vascular function and reduced nitric oxide (NO)-bioavailability have been recognized to be involved in aging and Alzheimer's Disease (AD). A non-pharmacological treatment that is gaining clinical interest in the context of vascular function is dietary inorganic nitrate (NO3-) supplementation which increases NO-bioavailability through the NO3- -nitrite (NO2-) - NO pathway. This treatment has been demonstrated to improve vascular function in several clinical populations, but no study has investigated the effects in individuals with AD. Therefore, changes in plasma NO3- and NO2- and vascular responsiveness (hyperemic response to single-passive leg movement (ΔPLM)) were measured in individuals with AD (n = 10, 76 ± 9 years), healthy elderly (OLD, n = 10, 75 ± 6 years), and young individuals (YN, n = 10, 25 ± 4 years) before (T0) and hourly for 4 h (T1, T2, T3, and T4) after ingestion of either NO3--rich beetroot juice (BR) or a placebo (PLA). No changes in NO3- and NO2-, nor ΔPLM were detected in any group following PLA intake. Plasma NO3- and NO2- increased significantly in all three groups at T1 (p < 0.001) and remained elevated for the rest of the trial. The same trend was found in ΔPLM, which significantly increased in all three groups over the time (p < 0.001). However, AD exhibited significantly lower ΔPLM values at any time point compared to YN (p < 0.001) and OLD (p < 0.001). These data suggest that AD-individuals included in this study were able to reduce NO3- to NO2- and to increase NO-mediated vascular responsiveness as non-AD-individuals. Other mechanisms, beyond NO-bioavailability, may be involved in vascular dysfunction in patients with AD. This research suggests that an acute administration of inorganic nitrate is not enough to revert chronically adapted vascular properties and completely restore vascular responsiveness in AD.
Collapse
Affiliation(s)
- Anna Pedrinolla
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento, Italy.
| | - Gianluigi Dorelli
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Simone Porcelli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Mia Burleigh
- Sport and Physical Activity Research Institute, University of the West of Scotland, Blantyre, Scotland, UK
| | - Martina Mendo
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Camilla Martignon
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Cristina Fonte
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Chris Easton
- Sport and Physical Activity Research Institute, University of the West of Scotland, Blantyre, Scotland, UK; School of Energy, Geoscience, Infrastructure and Society, Heriot-Watt University, Edinburgh, Scotland, UK
| | | | - Federico Schena
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Massimo Venturelli
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy; Department of Internal Medicine Section of Geriatrics, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
14
|
Knudsen LV, Michel TM, Farahani ZA, Vafaee MS. Multimodal neuroimaging insights into the neurobiology of healthy aging across the lifespan. Eur J Nucl Med Mol Imaging 2025; 52:2267-2278. [PMID: 39890633 PMCID: PMC12119650 DOI: 10.1007/s00259-025-07100-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025]
Abstract
PURPOSE The purpose of this study was to advance our understanding of the neurobiology of healthy aging, which is crucial for improving quality of life and preventing age-related diseases. Despite its importance, a comprehensive investigation of this process has yet to be fully characterized. METHODS We used a hybrid PET/MRI scanner to assess neurophysiological parameters in 80 healthy individuals aged 20-78. Cerebral amyloid-beta (Aβ) deposition and glucose metabolism were assessed using PET scans, while participants underwent simultaneous MRI scans. RESULTS We found a positive correlation between Aβ-deposition and aging, and a negative correlation between glucose metabolism and aging. The insula showed the strongest negative correlation between glucose metabolism and age (Spearman's r = -0.683, 95% CI [-0.79, -0.54], p < 0.0001), while the posterior cingulate cortex had the strongest positive correlation between Aβ-deposition and age (Spearman's r = 0.479, 95% CI [0.28, 0.64], p < 0.0001). These results suggest a spatially dependent link between Aβ-deposition and metabolism in healthy older adults, indicating a compensatory mechanism in early Alzheimer's. Additionally, Aβ-deposition was linked to changes in interregional neural communication. CONCLUSIONS Our study confirms previous findings on aging and offers new insights, particularly on the role of Aβ-deposition in healthy aging. We observed a linear increase in Aβ-deposition, alongside decreases in white matter integrity, cerebral blood flow, and glucose metabolism. Additionally, we identified a complex regional relationship between Aβ-deposition, glucose metabolism, and neural communication, possibly reflecting compensatory mechanisms.
Collapse
Affiliation(s)
- Laust Vind Knudsen
- Department of Psychiatry, University of Southern Denmark, Odense University Hospital, Odense C, 5000, Denmark
| | - Tanja Maria Michel
- Department of Psychiatry, University of Southern Denmark, Odense University Hospital, Odense C, 5000, Denmark
| | | | - Manouchehr Seyedi Vafaee
- Department of Psychiatry, University of Southern Denmark, Odense University Hospital, Odense C, 5000, Denmark.
| |
Collapse
|
15
|
Kordi R, Andrews TJ, Hicar MD. Infections, genetics, and Alzheimer's disease: Exploring the pathogenic factors for innovative therapies. Virology 2025; 607:110523. [PMID: 40174330 DOI: 10.1016/j.virol.2025.110523] [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: 01/10/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative condition that creates a significant global health challenge and profoundly affects patients and their families. Recent research has highlighted the critical role of microorganisms, particularly viral infections, in the pathogenesis of AD. The involvement of viral infections in AD pathogenesis is predominantly attributed to their ability to induce neuroinflammation and amyloid beta (Aβ) deposition in the brain. The extant research exploring the relationship between viruses and AD has focused largely on Herpesviridae family. Traces of Herpesviruses, such as Herpes Simplex Virus-1 and Epstein Barr Virus, have been found in the brains of patients with AD. These viruses are thought to contribute to the disease progression by triggering chronic inflammatory responses in the brain. They can remain dormant in the brain, and become reactivated due to stress, a secondary viral infection, or immune-senescence in older adults. This review focuses on the association between Herpesviridae and bacterial infections with AD. We explore the genetic factors that might regulate viral illness and discuss clinical trials investigating antiviral and anti-inflammatory agents as possible therapeutic strategies to mitigate cognitive decline in patients with AD. In summary, understanding the interplay between infections, genetic factors, and AD pathogenesis may pave the way for novel therapeutic approaches, facilitating better management and possibly even prevent this debilitating disease.
Collapse
Affiliation(s)
- Ramesh Kordi
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Ted J Andrews
- Department of Pediatrics, Division of Developmental Pediatrics and Rehabilitation, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Mark D Hicar
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
16
|
Limone A, Di Napoli C, Napolitano F, Imbò B, Minopoli G, Bagnoli S, Izzo A, Paladino S, Nacmias B, De Matteis MA, Montuori N, Lavecchia A, Sarnataro D. Targeting RPSA to modulate endosomal trafficking and amyloidogenesis in genetic Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167753. [PMID: 40037473 DOI: 10.1016/j.bbadis.2025.167753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
The "amyloid cascade hypothesis" for Alzheimer's disease (AD) pathogenesis, highlights the accumulation of amyloid-β (Aβ) as a crucial trigger for the pathology. However, AD is an extremely complex disease influenced by multiple pathophysiological processes, making it impossible to attribute its onset to a single hypothesis. The endocytic pathway, where the amyloidogenic processing of APP occurs, has emerged as a pathogenic "hub" for AD. In this study, we found altered homeostasis and dynamics of endolysosomal compartments in fibroblasts from patients affected by a genetic form of AD (APP V717I mutation). These alterations corresponded to an abnormal trafficking of APP along the endolysosomal pathway, favouring its amyloidogenic processing. The identification of APP interactors involved in its trafficking and processing, and finding molecules able to interfere with these interactions, represents a promising therapeutic approach. However, the role of endosomal pathway and the possibility of modulating APP processing through it remains elusive. Among the proteins participating to APP metabolism, the RPSA receptor and its inhibitor molecule NSC47924 have been identified. In this study, we found that the inhibitor, likely by displacing APP from interaction with its receptor, reduced APP accumulation in EEs in AD cells, finally restoring both endosomal dynamics and APP distribution to those of unaffected cells. We also demonstrated that RPSA inhibition affected the aberrant APP cleavage, as it reduced the production of both APP-βCTF (C-Terminal Fragment) and Aβ in AD fibroblasts. These results highlight significant differences in endolysosomal compartments and APP processing in AD-affected cells, refining our understanding of APP/RPSA intersection.
Collapse
Affiliation(s)
- Adriana Limone
- University of Naples "Federico II", Dept. of Molecular Medicine and Medical Biotechnology, Via S. Pansini 5, 80131 Naples, Italy
| | - Clelia Di Napoli
- University of Naples "Federico II", Dept. of Molecular Medicine and Medical Biotechnology, Via S. Pansini 5, 80131 Naples, Italy
| | - Filomena Napolitano
- University of Naples "Federico II"- Dept. of Translational Medical Sciences, Via S. Pansini 5, 80131 Naples, Italy
| | - Barbara Imbò
- University of Naples "Federico II", Dept. of Molecular Medicine and Medical Biotechnology, Via S. Pansini 5, 80131 Naples, Italy
| | - Giuseppina Minopoli
- University of Naples "Federico II", Dept. of Molecular Medicine and Medical Biotechnology, Via S. Pansini 5, 80131 Naples, Italy
| | - Silvia Bagnoli
- University of Florence, Dept. of Neuroscience, Psychology, Drug Research and Child Health, Viale Pieraccini 6, 50139 Florence, Italy
| | - Antonella Izzo
- University of Naples "Federico II", Dept. of Molecular Medicine and Medical Biotechnology, Via S. Pansini 5, 80131 Naples, Italy
| | - Simona Paladino
- University of Naples "Federico II", Dept. of Molecular Medicine and Medical Biotechnology, Via S. Pansini 5, 80131 Naples, Italy
| | - Benedetta Nacmias
- University of Florence, Dept. of Neuroscience, Psychology, Drug Research and Child Health, Viale Pieraccini 6, 50139 Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Maria Antonietta De Matteis
- University of Naples "Federico II", Dept. of Molecular Medicine and Medical Biotechnology, Via S. Pansini 5, 80131 Naples, Italy; Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, NA, Italy
| | - Nunzia Montuori
- University of Naples "Federico II"- Dept. of Translational Medical Sciences, Via S. Pansini 5, 80131 Naples, Italy
| | - Antonio Lavecchia
- University of Naples "Federico II"- Dept. of Pharmacy, "Drug Discovery Lab", Via D. Montesano 49, 80131, Naples, Italy
| | - Daniela Sarnataro
- University of Naples "Federico II", Dept. of Molecular Medicine and Medical Biotechnology, Via S. Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
17
|
Wang C, Zhang W, Ni M, Wang Q, Liu C, Dai L, Zhang M, Shen Y, Gao F. Deep-learning based multi-modal models for brain age, cognition and amyloid pathology prediction. Alzheimers Res Ther 2025; 17:126. [PMID: 40450379 DOI: 10.1186/s13195-025-01773-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 05/23/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND Magnetic resonance imaging (MRI), combined with artificial intelligence techniques, has improved our understanding of brain structural change and enabled the estimation of brain age. Neurodegenerative disorders, such as Alzheimer's disease (AD), have been linked to accelerated brain aging. In this study, we aimed to develop a deep-learning framework that processes and integrates MRI images to more accurately predict brain age, cognitive function, and amyloid pathology. METHODS In this study, we aimed to develop a deep-learning framework that processes and integrates MRI images to more accurately predict brain age, cognitive function, and amyloid pathology.We collected over 10,000 T1-weighted MRI scans from more than 7,000 individuals across six cohorts. We designed a multi-modal deep-learning framework that employs 3D convolutional neural networks to analyze MRI and additional neural networks to evaluate demographic data. Our initial model focused on predicting brain age, serving as a foundational model from which we developed separate models for cognition function and amyloid plaque prediction through transfer learning. RESULTS The brain age prediction model achieved the mean absolute error (MAE) for cognitive normal population in the ADNI (test) datasets of 3.302 years. The gap between predicted brain age and chronological age significantly increases while cognition declines. The cognition prediction model exhibited a root mean square error (RMSE) of 0.334 for the Clinical Dementia Rating (CDR) regression task, achieving an area under the curve (AUC) of approximately 0.95 in identifying ing dementia patients. Dementia related brain regions, such as the medial temporal lobe, were identified by our model. Finally, amyloid plaque prediction model was trained to predict amyloid plaque, and achieved an AUC about 0.8 for dementia patients. CONCLUSIONS These findings indicate that the present predictive models can identify subtle changes in brain structure, enabling precise estimates of brain age, cognitive status, and amyloid pathology. Such models could facilitate the use of MRI as a non-invasive diagnostic tool for neurodegenerative diseases, including AD.
Collapse
Affiliation(s)
- Chenxi Wang
- Department of International Medical, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Weiwei Zhang
- Department of International Medical, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ming Ni
- Department of Nuclear Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Qiong Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Chang Liu
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Linbin Dai
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Mengguo Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Yong Shen
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China.
| | - Feng Gao
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
18
|
Li X, Wu Z, Si X, Li J, Wu G, Wang M. The role of mitochondrial dysfunction in the pathogenesis of Alzheimer's disease and future strategies for targeted therapy. Eur J Med Res 2025; 30:434. [PMID: 40450332 DOI: 10.1186/s40001-025-02699-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: 01/13/2025] [Accepted: 05/17/2025] [Indexed: 06/03/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive cognitive decline, behavioral impairments, and psychiatric comorbidities. The pathogenesis of AD remains incompletely elucidated, despite advances in dominant hypotheses such as the β-amyloid (Aβ) cascade, tauopathy, cholinergic deficiency, and neuroinflammation mechanisms. However, these hypotheses inadequately explain the multifactorial nature of AD, which exposes limitations in our understanding of its mechanisms. Mitochondrial dysfunction is known to play a pivotal role in AD, and since patients exhibit intracellular mitochondrial dysfunction and structural changes in the brain at an early stage, correcting the imbalance of mitochondrial homeostasis and the cytopathological changes caused by it may be a potential target for early treatment of AD. Mitochondrial structural abnormalities accelerate AD pathogenesis. For instance, structural and functional alterations in the mitochondria-associated endoplasmic reticulum membrane (MAM) can disrupt intracellular Ca2⁺ homeostasis and cholesterol metabolism, consequently promoting Aβ accumulation. In addition, the overaccumulation of Aβ and hyperphosphorylated tau proteins can further damage neurons by disrupting mitochondrial integrity and mitophagy, thereby amplifying pathological aggregation and exacerbating neurodegeneration in AD. Furthermore, Aβ deposition and abnormal tau proteins can disrupt mitochondrial dynamics through dysregulation of fission/fusion proteins, leading to excessive mitochondrial fragmentation and subsequent dysfunction. Additionally, hyperphosphorylated tau proteins can impair mitochondrial transport, resulting in axonal dysfunction in AD. This article reviews the biological significance of mitochondrial structural morphology, dynamics, and mitochondrial DNA (mtDNA) instability in AD pathology, emphasizing mitophagy abnormalities as a critical contributor to AD progression. Additionally, mitochondrial biogenesis and proteostasis are critical for maintaining mitochondrial function and integrity. Impairments in these processes have been implicated in the progression of AD, further highlighting the multifaceted role of mitochondrial dysfunction in neurodegeneration. It further discusses the therapeutic potential of mitochondria-targeted strategies for AD drug development.
Collapse
Affiliation(s)
- Xin Li
- Department of Neurology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Ziyang Wu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xiaying Si
- Department of Psychiatry, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jing Li
- Department of Neurology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Guode Wu
- Department of Neurology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Manxia Wang
- Department of Neurology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China.
| |
Collapse
|
19
|
Frederiksen KS, Boada M, Dubois B, Engelborghs S, Frisoni GB, Georges J, Hort J, Jönsson L, Kramberger MG, Ousset PJ, Scarmeas N, Schmidt R, Schott JM, Spiro L, Waldemar G, Winblad B, Jessen F, Frölich L. Navigating the introduction of anti-amyloid therapy in Europe: a position statement by individual members of the EADC. Alzheimers Res Ther 2025; 17:116. [PMID: 40413561 PMCID: PMC12102950 DOI: 10.1186/s13195-025-01766-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 05/13/2025] [Indexed: 05/27/2025]
Abstract
INTRODUCTION Anti-amyloid antibodies for the treatment of Alzheimer´s disease (AD) are currently being evaluated for approval and reimbursement in Europe. An approval brings opportunities, but also challenges to health care systems across Europe. The objective of this position paper is to provide guidance from experts in the field in terms of navigating implementation. METHODS Members of the European Alzheimer's Disease Consortium and a representative of Alzheimer Europe convened to formulate recommendations covering key areas related to the possible implementation of anti-amyloid antibodies in AD through online discussions and 2 rounds of online voting with an 80% threshold for a position to be accepted. RESULTS In total, 24 recommendations were developed covering the research landscape and priorities within research in AD following a possible approval, potential impact on health care systems and diagnostic pathways, and communication to patients about anti-amyloid antibodies. Anti-amyloid antibodies are regarded as a substantial innovation with an important clinical impact. In addition, however, new compounds with other mechanisms of action and/or route of administration are also needed. Approval of new treatments will require changes to existing patient pathways and real-world data needs to be generated. CONCLUSION Comprehensive guidance is provided on the potential implementation of anti-amyloid antibody therapies in Europe following possible approval. Emphasis is placed on the necessity of regularly updating recommendations as new evidence emerges in the coming years.
Collapse
Affiliation(s)
- Kristian S Frederiksen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Mercé Boada
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Bruno Dubois
- Institute of Memory and Alzheimer's Disease (IM2 A), Department of Neurology, Assistance Publique-Hôpitaux de Paris (AP-HP), Sorbonne University, Pitié-Salpêtrière Hospital, Paris, France
- Frontlab, Paris Brain Institute (Institut du Cerveau, ICM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Sebastiaan Engelborghs
- Department of Neurology and Bru-BRAIN, NEUR Research Group, Center for Neurosciences (C4 N), Universitair Ziekenhuis Brusseland, VrijeUniversiteit Brussel (VUB) , Brussels, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Giovanni B Frisoni
- LANVIE - Laboratory of Neuroimaging of Aging, Memory Centerand, University Hospitals and University of Geneva , Geneva, Switzerland
| | | | - Jakub Hort
- Department of Neurology, Second Faculty of Medicine, Memory Clinic, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Linus Jönsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Milica G Kramberger
- Department of Neurology, Faculty of Medicine, University Medical Center, University of Ljubljana, Ljubljana, Slovenia
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Pierre-Jean Ousset
- Toulouse Clinical Research Center, Toulouse University Hospital, Toulouse, France
| | - Nikolaos Scarmeas
- 1, Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School GR, Athens, Greece
- Department of Neurology, Taub Institute for Research in Alzheimer's Disease and the Aging Brain, The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University Graz, Graz, Austria
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, UCL, London, UK
| | - Luiza Spiro
- Carol Davila" University of Medicine and Pharmacy (www.umfcd.ro), Bucharest, Romania
- Faculty, Geneva College of Longevity Science (www.gcls.study), Geneva, Switzerland
- Faculty, Geneva School of Business Administration, University of Applied Sciences of Western Switzerland (www.hesge.ch), Geneva, Switzerland
- President "Ana ASLAN International" Foundation (www.anaaslanacademy.ro), Excellence Memory Center, Brain Health, and Longevity Medicine, Bucharest, Romania
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bengt Winblad
- Aging and Inflammation Theme, Karolinska University Hospital, Stockholm, Sweden
- Division of Neurogeriatrics, Dept of Neurobiology, Care Sciences and Society, Theme Inflammation & Aging, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Frank Jessen
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| |
Collapse
|
20
|
Ma'u E, Cullum S, Röhr S, Brayne EC. Are modifiable risk factors for dementia really modifiable? Curr Opin Psychiatry 2025:00001504-990000000-00177. [PMID: 40423572 DOI: 10.1097/yco.0000000000001018] [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] [Indexed: 05/28/2025]
Abstract
PURPOSE OF REVIEW The 2024 Lancet Commission estimates 45% of dementias worldwide are preventable if 14 potentially modifiable risk factors for dementia were eliminated. While this is unlikely, there is evidence that even modest risk factor reduction will have significant benefits. Whether this is best achieved at the level of the individual or broader population level approaches is the purpose of this review. RECENT FINDINGS To date, evidence for the efficacy of individual-level interventions in preventing cognitive decline or dementia is modest at best. Reasons for this include the sociodemographic and risk profile of study participants and complex disease causes, while overlooking the underlying social and commercial determinants of health influencing risk exposure. There is, however, growing evidence supporting population-level approaches to dementia risk reduction. Trend studies from high-income countries showing declines in dementia incidence over recent decades suggest their effectiveness. SUMMARY The limited evidence for the efficacy, let alone effectiveness, of individual-level interventions is in part because they operate within the influence of social and commercial determinants of health. For significant and sustained risk factor reduction, population-level interventions targeting the underlying determinants of risk factor exposure across the life course, with sensitivity to diverse contexts, are required.
Collapse
Affiliation(s)
- Etuini Ma'u
- Department of Psychological Medicine, University of Auckland, Auckland
- Te Whatu Ora Waikato, Hamilton, New Zealand
| | - Sarah Cullum
- Department of Psychological Medicine, University of Auckland, Auckland
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
| | - Susanne Röhr
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Centre for Healthy Brain Ageing (CHeBA), School of Clinical Medicine, UNSW Sydney, Sydney, Australia
| | | |
Collapse
|
21
|
Chen J, Zhu Z, Xu Y. Signs of Alzheimer's Disease: Tied to Aging. Int J Mol Sci 2025; 26:4974. [PMID: 40507786 PMCID: PMC12154111 DOI: 10.3390/ijms26114974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025] Open
Abstract
: Alzheimer's disease (AD) is a neurodegenerative disorder closely associated with aging, and its pathogenesis involves the interaction of multidimensional pathophysiologic processes. This review outlines the core mechanisms linking aging and AD. The amyloid cascade hypothesis emphasizes that abnormal deposition of amyloid-β (Aβ) triggers neuronal damage and synaptic dysfunction, which is exacerbated by aging-associated declines in protein clearance. Neuroinflammation, a synergistic pathogenetic factor in AD, is mediated by microglia activation, creating a vicious cycle with Aβ and tau pathology. The cholinergic hypothesis states that the degeneration of cholinergic neurons in the basal forebrain can lead to acetylcholine deficiency, which is directly associated with cognitive decline. Endothelial disorders promote neuroinflammation and metabolic waste accumulation through blood-brain barrier dysfunction and cerebral vascular abnormalities. In addition, glutamate-mediated excitotoxicity and mitochondrial dysfunction (e.g., oxidative stress and energy metabolism imbalance) further lead to neuronal death, and aging-associated declines in mitochondrial autophagy exacerbate such damage. This review also explores the application of animal models that mimic AD and aging in studying these mechanisms and summarizes therapeutic strategies targeting these pathways. Future studies need to integrate multi-targeted therapies and focus on the role of the aging microenvironment in regulating AD pathology in order to develop more effective early diagnosis and treatment options.
Collapse
Affiliation(s)
| | | | - Yuanyuan Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Science, Jilin University, Changchun 130062, China; (J.C.); (Z.Z.)
| |
Collapse
|
22
|
Ikebara JM, Jorge RS, Marinho LSR, Higa GSV, Adhikari A, Reis FMCV, Borges FS, Ulrich H, Takada SH, De Pasquale R, Kihara AH. Hippocampal Interneurons Shape Spatial Coding Alterations in Neurological Disorders. Mol Neurobiol 2025:10.1007/s12035-025-05020-2. [PMID: 40392508 DOI: 10.1007/s12035-025-05020-2] [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: 09/19/2024] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Hippocampal interneurons (INs) play a fundamental role in regulating neural oscillations, modulating excitatory circuits, and shaping spatial representation. While historically overshadowed by excitatory pyramidal cells in spatial coding research, recent advances have demonstrated that inhibitory INs not only coordinate network dynamics but also contribute directly to spatial information processing. This review aims to provide a novel integrative perspective on how distinct IN subtypes participate in spatial coding and how their dysfunction contributes to cognitive deficits in neurological disorders such as epilepsy, Alzheimer's disease (AD), traumatic brain injury (TBI), and cerebral hypoxia-ischemia. We synthesize recent findings demonstrating that different IN classes-including parvalbumin (PV)-, somatostatin (SST)-, cholecystokinin (CCK)-, and calretinin (CR)-expressing neurons-exhibit spatially selective activity, challenging traditional views of spatial representation, and influence memory consolidation through network-level interactions. By leveraging cutting-edge techniques such as in vivo calcium imaging and optogenetics, new evidence suggests that INs encode spatial information with a level of specificity previously attributed only to pyramidal cells. Furthermore, we investigate the impact of inhibitory circuit dysfunction in neurological disorders, examining how disruptions in interneuronal activity lead to impaired theta-gamma coupling, altered sharp wave ripples, and destabilized place cell representations, ultimately resulting in spatial memory deficits. This review advances the field by shifting the focus from pyramidal-centered models to a more nuanced understanding of the hippocampal network, emphasizing the active role of INs in spatial coding. By highlighting the translational potential of targeting inhibitory circuits for therapeutic interventions, we propose novel strategies for restoring hippocampal network function in neurological conditions. Readers will gain a comprehensive understanding of the emerging role of INs in spatial representation and the critical implications of their dysfunction, paving the way for future research on interneuron-targeted treatments for cognitive disorders.
Collapse
Affiliation(s)
- Juliane Midori Ikebara
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Renata Silva Jorge
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Luciana Simões Rafagnin Marinho
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Guilherme Shigueto Vilar Higa
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Avishek Adhikari
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Fernando M C V Reis
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Fernando S Borges
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY, 11203, USA
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil
| | - Roberto De Pasquale
- Neurophysiology Laboratory, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Alexandre Hiroaki Kihara
- Neurogenetics Laboratory, Center of Mathematics, Computation and Cognition, Federal University of ABC (UFABC), Alameda da Universidade, S/N, São Bernardo Do Campo, SP, 09606-045, Brazil.
| |
Collapse
|
23
|
Campagna J, Jagodzinska B, Wi D, Zhu C, Lee J, Cohn W, Jun M, Elias C, Padder S, Descamps O, Peters-Libeu C, Zhang Q, Gorostiza O, Poksay K, Spilman P, Bredesen D, John V. Discovery of an APP-selective BACE1 inhibitor for Alzheimer's disease. Neurotherapeutics 2025:e00610. [PMID: 40399225 DOI: 10.1016/j.neurot.2025.e00610] [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: 11/15/2024] [Revised: 04/16/2025] [Accepted: 05/06/2025] [Indexed: 05/23/2025] Open
Abstract
Inhibition of amyloid precursor protein (APP) beta-site cleaving enzyme 1 (BACE1) has been a target for Alzheimer's disease (AD) therapeutic development. Here, we report our identification of APP-selective BACE1 (ASBI) inhibitors that are selective for APP as the substrate and BACE1 as the target enzyme. A known fluoro aminohydantoin (FAH) inhibitor compound was identified by screening a compound library for inhibition of BACE1 cleavage of a maltose binding protein (MBP)-conjugated-APPC125 substrate followed by optimization and IC50 determination using the P5-P5' activity assay. Optimization of the screening hit led to candidate FAH65, which displays selectivity for inhibition of APP cleavage with little activity against other BACE1 substrates neuregulin 1 (NRG1) or p-selectin glycoprotein ligand-1 (PSGL1). FAH65 shows little inhibitory activity against other aspartyl proteases cathepsin D (Cat D) and BACE2. FAH65 reduces BACE1 cleavage products soluble APPβ (sAPPβ) and the β C-terminal fragment (βCTF), as well as amyloid-β (Aβ) 1-40 and 1-42, both in vitro in cells and in vivo in an animal model of AD. In a murine model of AD, FAH65 improved the discrimination score in the Novel Object Recognition (NOR) memory testing paradigm. The active enantiomer of racemate FAH65, FAH65E(-), displays good brain-penetrance and target engagement, meriting further pre-clinical development as an ASBI that may reduce Aβ levels and overcome the deleterious effects of the non-selective BACE1 inhibitors that have failed in the clinic. FAH65E(-) has the potential to be a first-in-class oral therapy that could be used in conjunction with an approved anti-Aβ antibody therapy for AD.
Collapse
Affiliation(s)
- Jesus Campagna
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA
| | - Barbara Jagodzinska
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA
| | - Dongwook Wi
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA
| | - Chunni Zhu
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA
| | - Jessica Lee
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA
| | - Whitaker Cohn
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA
| | - Michael Jun
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA
| | - Chris Elias
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA
| | - Samar Padder
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA
| | | | | | - Qiang Zhang
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | - Karen Poksay
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Patricia Spilman
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA
| | - Dale Bredesen
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA; Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Varghese John
- Drug Discovery Lab (DDL), Department of Neurology, Easton Center for Alzheimer's Disease Research and Care, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA, 90095, USA.
| |
Collapse
|
24
|
Jin Y, Ma X, Liu S, Zong S, Cheng Y, Zhang H, Wang C, Li Y. Application of Natural Products in Neurodegenerative Diseases by Intranasal Administration: A Review. Pharmaceutics 2025; 17:675. [PMID: 40430965 PMCID: PMC12114702 DOI: 10.3390/pharmaceutics17050675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Natural products derived from traditional Chinese medicine have received significant attention as potential treatments for neurodegenerative disorders due to their wide availability, demonstrated efficacy, and favorable safety profiles. Intranasal delivery provides distinct advantages for targeting the central nervous system (CNS), enabling direct therapeutic agent delivery to the brain by bypassing the blood-brain barrier (BBB). This review evaluates natural products administered intranasally for neurodegenerative diseases (NDs), highlighting their therapeutic potential and addressing formulation challenges related to physicochemical properties. Strategic optimization approaches are proposed, including novel carrier systems, molecular modifications, and combination therapies. By discussing current difficulties and offering practical recommendations, this review aims to encourage further scholarly research and clinical application.
Collapse
Affiliation(s)
- Yu Jin
- Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (Y.J.)
| | - Xinyu Ma
- Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (Y.J.)
| | - Shuo Liu
- Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (Y.J.)
| | - Shiyu Zong
- Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi’an 710001, China
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an 710001, China
| | - Yunlong Cheng
- Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi’an 710001, China
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an 710001, China
| | - Hong Zhang
- Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (Y.J.)
- Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi’an 710001, China
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an 710001, China
| | - Chunliu Wang
- Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi’an 710001, China
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an 710001, China
| | - Ye Li
- Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (Y.J.)
- Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi’an 710001, China
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an 710001, China
| |
Collapse
|
25
|
Lu F, Li L, Zheng B, Wang C, Liu Z, Huang X, Song L, Ding C, Li Y. Icariin alleviates cognitive dysfunction by reducing neuroinflammation via the cGAS-STING pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 350:120010. [PMID: 40403897 DOI: 10.1016/j.jep.2025.120010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 05/18/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Epimedium brevicornu Maxim, a Chinese herbal medicine, is known for its efficacy in nourishing the kidneys. Icariin (ICA), the primary active ingredient in Epimedium brevicornu Maxim., possesses multiple pharmacological properties, yet its impact on Alzheimer's disease (AD) warrants further exploration. AIM OF THE STUDY Study aims to explore the inhibitory impact of ICA on neuroinflammation in AD via the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. MATERIALS AND METHODS SPF-grade male ICR mice were used to establish an AD model by lateral ventricle injection of Aβ1-42. Behavioral, pathological assessments, as well as immunofluorescence staining, molecular docking, and Western blot analyses, were conducted to evaluate the effects of ICA treatment on memory function, neuronal damage, neuroinflammation, and the cGAS- STING pathway in mice. RESULTS ICA significantly improved memory impairment, alleviated neuronal damage and apoptosis, and suppressed neuroinflammation in AD mice. Additionally, ICA inhibited microglial hyperactivation, promoting the transition from the M1 to the M2 phenotype. It specifically inhibited the activation of the cGAS-STING pathway and down-regulated the expression of cGAS, STING, p-TBK1/TBK1, p-IRF3/IRF3 and p-NF-κB/NF-κB. Furthermore, molecular docking revealed that the binding energy between ICA and cGAS was -7.07 kcal/mol, indicating a stable interaction. Further validation using the cGAS-selective small molecule inhibitor RU.521 confirmed the protective effects of ICA against cGAS-STING signaling on microglial transformation and neuroinflammation. CONCLUSION ICA exhibits therapeutic potential in AD by inhibiting microglial transformation and neuroinflammation through the cGAS-STING pathway, positioning it as a candidate drug for AD treatment targeting this pathway.
Collapse
Affiliation(s)
- Fengjuan Lu
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, China
| | - Lu Li
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, China; Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Bilian Zheng
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, China
| | - Ce Wang
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, China
| | - Ziying Liu
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, China
| | - Xingshuo Huang
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, China
| | - Linyue Song
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, China
| | - Chong Ding
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, China
| | - Yang Li
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, China.
| |
Collapse
|
26
|
Gillespie NA, Neale MC, Panizzon MS, McKenzie RE, Tu XM, Xian H, Reynolds CA, Lyons MJ, Rissman RA, Elman JA, Franz C, Kremen WS. Testing the causal impact of plasma amyloid on total Tau using a genetically informative sample of adult male twins. AGING BRAIN 2025; 7:100139. [PMID: 40487291 PMCID: PMC12145560 DOI: 10.1016/j.nbas.2025.100139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 06/18/2025] Open
Abstract
The amyloid cascade hypothesis predicts that amyloid-beta (Aβ) aggregation drives tau tangle accumulation. We tested competing causal and non-causal hypotheses regarding the direction of causation between Aβ40 and Aβ42 and total Tau (t-Tau) plasma biomarkers. Plasma Aβ40, Aβ42, t-Tau, and neurofilament light chain (NFL) were measured in 1,035 men (mean = 67.0 years) using Simoa immunoassays. Genetically informative twin modeling tested the direction of causation between Aβs and t-Tau. No clear evidence that Aβ40 or Aβ42 directly causes t-Tau was observed. Instead, the alternative causal hypotheses also fit the data well. In contrast, exploratory analyses suggested a causal impact of the Aβ biomarkers on NFL. Separately, reciprocal causation was observed between t-Tau and NFL. Plasma Aβ40 or Aβ42 do not appear to have a direct causal impact on t-Tau, though our use of total rather than phosphorylated tau was a limitation. In contrast, Aβ biomarkers appeared to causally impact NFL in cognitively unimpaired men in their late 60 s.
Collapse
Affiliation(s)
- Nathan A. Gillespie
- Virginia Institute for Psychiatric and Behaviour Genetics, Department of Psychiatry, Virginia Commonwealth University, Box 980126, Richmond, VA 23298-0126, USA
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4006, Australia
| | - Michael C. Neale
- Virginia Institute for Psychiatric and Behaviour Genetics, Department of Psychiatry, Virginia Commonwealth University, Box 980126, Richmond, VA 23298-0126, USA
| | - Matthew S. Panizzon
- Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Psychology, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
| | - Ruth E. McKenzie
- Winston School of Education and Social Policy at Merrimack College, 315 Turnpike Street, North Andover, MA 01845, USA
- Department of Family Medicine and Public Health, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Xin M. Tu
- Department of Psychology, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
- Department of Epidemiology and Biostatistics, Saint Louis University, 3545 Lafayette Ave, St. Louis, MO 63104, USA
| | - Hong Xian
- Research Service, VA St. Louis Healthcare System, 1 Jefferson Barracks Drive, St. Louis, MO 63125, USA
- Institute for Behavioral Genetics and Department of Psychology and Neuroscience, University of Colorado, Boulder, 1480 30th Street, Boulder, CO 80309, USA
| | - Chandra A. Reynolds
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
| | - Michael J. Lyons
- Sam and Rose Stein Institute for Research on Aging, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Robert A. Rissman
- Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Psychology, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
| | - Jeremy A. Elman
- Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Psychology, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
| | - Carol Franz
- Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Psychology, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
| | - William S. Kremen
- Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Psychology, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
| |
Collapse
|
27
|
Xing S, Kang X, Wang R, Wang C, Wang Y, Bao X, Zhao J. Microbial Production of Nicotinamide Mononucleotide: Key Enzymes Discovery, Host Cells Selection, and Pathways Design and Optimization. ACS Synth Biol 2025; 14:1352-1366. [PMID: 40237164 DOI: 10.1021/acssynbio.5c00038] [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: 04/18/2025]
Abstract
As an important bioactive substance in cells, nicotinamide mononucleotide (NMN) has been proven to play an important role in antiaging, treatment of neurodegenerative diseases, and cardioprotection. It presents a high potential for application in the research fields of functional foods, cosmetics, healthcare products, and active pharmaceuticals. With the increased demand, whether NMN can achieve large-scale industrial production has been a wide concern. The chemical synthesis method of NMN mainly faces the problems of separation, purification, and complex process control; in contrast, biosynthesis methods such as microbial fermentation and enzyme catalysis are considered to be the mainstream of the future industrial production of NMN due to the advantages of environmental friendliness, high efficiency, and simple separation. This review first describes the physiological functions of NMN and the related areas of its applications. Subsequently, it focuses on the research progress on different synthetic pathways of NMN in biosynthetic approaches, mining and modification of key enzymes, chassis cell design and optimization, and whole-cell catalysis. Meanwhile, the regulatory strategies, methods, and process control of the microbial synthesis of NMN are also elaborated, and the synthesis efficiencies of different chassis cells are systematically compared. Finally, this review summarizes the existing problems and challenges of microbial synthesis of NMN and proposes future strategies and directions to address these issues. This work provides technical references and a theoretical basis for researching efficient NMN microbial synthesis and application.
Collapse
Affiliation(s)
- Shuyi Xing
- State Key Laboratory of Green Papermaking and Resource Recycling, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Xiulong Kang
- State Key Laboratory of Green Papermaking and Resource Recycling, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Rui Wang
- State Key Laboratory of Green Papermaking and Resource Recycling, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Chengqiang Wang
- College of Life Sciences, National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, Shandong Key Laboratory of Agricultural Microbiology, Shandong Agricultural University, Taian 271018, China
| | - Yanjun Wang
- National Key Laboratory of Efficient Utilization of Nutrient Resources, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Xiaoming Bao
- State Key Laboratory of Green Papermaking and Resource Recycling, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Jianzhi Zhao
- State Key Laboratory of Green Papermaking and Resource Recycling, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| |
Collapse
|
28
|
Losinski GM, Key MN, Vidoni ED, Clutton J, Morris JK, Burns JM, Watts A. APOE4 and chronic health risk factors are associated with sex-specific preclinical Alzheimer's disease neuroimaging biomarkers. Front Glob Womens Health 2025; 6:1531062. [PMID: 40444147 PMCID: PMC12119584 DOI: 10.3389/fgwh.2025.1531062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Two thirds of Alzheimer's disease (AD) patients are female. Genetic and chronic health risk factors for AD affect females more negatively compared to males. Objective This multimodal neuroimaging study aimed to examine sex differences in cognitively unimpaired older adults on: (1) amyloid-β via 18F-AV-45 Florbetapir PET imaging, (2) neurodegeneration via T1 weighted MRI volumetrics, (3) cerebral blood flow via ASL-MRI. We identified AD risk factors including genetic (APOE genotype status) and health markers (fasting glucose, mean arterial pressure, waist-to-hip ratio, and android and gynoid body fat) associated with neuroimaging outcomes for which we observed sex differences. Methods Participants were sedentary, amyloid-β positive older adults (N = 112, ages 65-87 years) without evidence of cognitive impairment (CDR = 0). Results Multivariate analysis of covariance models adjusted for intracranial volume, age, and years of education demonstrated lower volume [F (7, 102) = 2.67, p = 0.014] and higher blood flow F (6, 102) = 4.25, p ≤ 0.001) among females compared to males in regions of interest connected to AD pathology and the estrogen receptor network. We did not observe sex differences in amyloid-β levels. Higher than optimal waist to hip ratio was most strongly associated with lower volume among female participants. Discussion Findings suggest genetic and chronic health risk factors are associated with sex-specific AD neuroimaging biomarkers. Underlying sex-specific biological pathways may explain these findings. Our results highlight the importance of considering sex differences in neuroimaging studies and when developing effective interventions for AD prevention and risk reduction.
Collapse
Affiliation(s)
- Genna M. Losinski
- Department of Psychology, University of Kansas, Lawrence, KS, United States
| | - Mickeal N. Key
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Eric D. Vidoni
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Jonathan Clutton
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Jill K. Morris
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Jeffrey M. Burns
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Amber Watts
- Department of Psychology, University of Kansas, Lawrence, KS, United States
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| |
Collapse
|
29
|
Kaewsaen R, Chanput WP, Rojanathammanee L, Golovko SA, Seeger DR, Golovko MY, Nookala S, Combs CK. Dietary Supplementation of Edible Mushroom Phallus atrovolvatus Aqueous Extract Attenuates Brain Changes in the AppNL-G-F Mouse Model of Alzheimer's Disease. Nutrients 2025; 17:1677. [PMID: 40431417 PMCID: PMC12114250 DOI: 10.3390/nu17101677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive dementia and brain accumulation of Aβ-peptide-containing plaques, gliosis, neuroimmune changes, and neurofibrillary tangles. Mushroom polysaccharides have been previously reported to have anti-neuroinflammation activity through the gut-brain axis. This study aimed to evaluate whether a dietary intervention with Phallus atrovolvatus, a recently identified edible mushroom in Thailand, could have a benefit on gut health and alleviate AD-related changes. Methods: Male and female 6-8-month-old littermate wild-type control (C57BL/6J) and AppNL-G-F mice were randomly assigned to either a control diet or a diet supplemented with mushroom aqueous extract (MAE) for 8 weeks to quantify changes in body weight, intestine, immune cells, short chain fatty acids, brain cytokines, amyloid-β (Aβ) levels, gliosis, and memory. Results: MAE had no adverse effects on gut leakiness and increased pyruvate levels in serum. Splenocyte immune profiling revealed a significant increase in the frequency of IgM+, IA_IE+, and CD14+ cells in MAE-administered AppNL-G-Ffemale mice compared to their vehicle controls. AppNL-G-Fmale mice that received MAE showed a significant increase in the frequency of cytotoxic CD8 T cells within the cervical lymph nodes compared to their wild-type counterparts. Aβ deposition and gliosis were significantly reduced in the hippocampi of the MAE-supplemented AppNL-G-F groups. However, MAE feeding did not alter spatial recognition memory in either sex or genotype compared to their vehicle groups. Conclusions: Our findings demonstrated that the administration of P. atrovolvatus aqueous extract showed neuroprotective potential against AD-related changes in the brain with no adverse impact on gut health and memory.
Collapse
Affiliation(s)
- Raweephorn Kaewsaen
- Department of Food Science and Technology, Faculty of Agro-Industry, Kasetsart University, 50 Ngam Wong Wan Rd., Ladyao, Chatuchak, Bangkok 10900, Thailand; (R.K.); (W.P.C.)
| | - Wasaporn Preteseille Chanput
- Department of Food Science and Technology, Faculty of Agro-Industry, Kasetsart University, 50 Ngam Wong Wan Rd., Ladyao, Chatuchak, Bangkok 10900, Thailand; (R.K.); (W.P.C.)
| | - Lalida Rojanathammanee
- School of Sports Science, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand;
| | - Svetlana A. Golovko
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.G.); (D.R.S.); (M.Y.G.); (S.N.)
| | - Drew R. Seeger
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.G.); (D.R.S.); (M.Y.G.); (S.N.)
| | - Mikhail Y. Golovko
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.G.); (D.R.S.); (M.Y.G.); (S.N.)
| | - Suba Nookala
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.G.); (D.R.S.); (M.Y.G.); (S.N.)
| | - Colin K. Combs
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.G.); (D.R.S.); (M.Y.G.); (S.N.)
| |
Collapse
|
30
|
Schultz B, Bobermin L, Menezes L, Gayger-Dias V, Da Silva VF, Taday J, Alves J, Tramontina F, Zanotto C, Dalmaz C, Leite MC, Leal RB, Quincozes-Santos A, Gonçalves CA. Sex-Dependent Changes in the Gene Expression of UPR-Associated Calcium-Binding Proteins in the STZ-Induced Model of Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-05046-6. [PMID: 40372671 DOI: 10.1007/s12035-025-05046-6] [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: 12/03/2024] [Accepted: 05/06/2025] [Indexed: 05/16/2025]
Abstract
Although the causes of Alzheimer's disease (AD) are still unknown, the unfolded protein response (UPR) is considered the basis for the pathogenesis of many degenerative diseases, including AD. The incidence of AD is slightly higher in the female population; however, biases continue to raise questions as to whether gender is a risk factor for the disease, as insulin resistance is. In this study, we used a sporadic model of Alzheimer's disease, induced by intracerebroventricularly-administered streptozotocin (STZ) in Wistar rats, to evaluate potential modulations in proteins involved in the UPR and the dependence of alterations on the sex of the animals. The rats were evaluated at two time points; 4 and 16 weeks post-STZ. At 16 weeks, cognitive deficit was observed in all rats treated with STZ, as well as an increase in glial fibrillary acid protein (GFAP), and a reduction in synaptophysin in the hippocampus. However, at 4 weeks, cognitive deficit was found only in males, in association with a reduction in synaptophysin. With regard to neurochemical changes in the AD model of STZ, we found sex-dependent differences in the gene expression of OASIS (an ATF-6-like UPR sensor in astrocytes), calpastatin (inhibitor protein of calpain 1/2), calpain-10, calcineurin, sorcin and CHOP. Taken together, results obtained herein contribute to the understanding of the pathogenesis of AD and indicate that the STZ-triggered UPR observed may be sex-dependent.
Collapse
Affiliation(s)
- Bruna Schultz
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil
| | - Larissa Bobermin
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil
| | - Leonardo Menezes
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil
| | - Vitor Gayger-Dias
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil
| | - Vanessa-Fernanda Da Silva
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil
| | - Jéssica Taday
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil
| | - Joelma Alves
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil
| | | | - Caroline Zanotto
- Biochemistry Laboratory, Grupo Hospitalar Conceição, Porto Alegre, RS, Brazil
| | - Carla Dalmaz
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil
- Graduate Program in Neurosciences, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
| | - Marina Concli Leite
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil
| | - Rodrigo Bainy Leal
- Graduate Program Biochemistry, Departament of Biochemistry, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - André Quincozes-Santos
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil
- Graduate Program in Neurosciences, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
| | - Carlos-Alberto Gonçalves
- Graduate Program in Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos Street 2600 Biochemistry Building Laboratory 33, Porto Alegre, RS, 90035-003, Brazil.
- Graduate Program in Neurosciences, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
31
|
Chen YH, Wang ZB, Liu XP, Mao ZQ. Cerebrospinal Fluid CCL25 as a Biomarker for Alzheimer's Disease: Associations with Pathology, Neurodegeneration, and Cognitive Decline. Mol Neurobiol 2025:10.1007/s12035-025-05007-z. [PMID: 40366557 DOI: 10.1007/s12035-025-05007-z] [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: 10/28/2024] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
Neuroinflammation plays a crucial role in Alzheimer's disease (AD) pathogenesis. We investigated the relationship between cerebrospinal fluid (CSF) C-C chemokine ligand 25 (CCL25), an inflammatory regulator, and AD pathology and progression. We analyzed data on CSF CCL25, AD biomarkers (CSF β-amyloid [Aβ]42, phosphorylated tau [pTau]181, amyloid positron emission tomography [PET]), postmortem neuropathology, magnetic resonance imaging-based neurodegeneration, and cognitive function from 703 participants in the Alzheimer's Disease Neuroimaging Initiative cohort. We found that elevated CSF CCL25 levels were associated with cognitive impairment, abnormal Aβ and tau pathology, greater brain atrophy, and worse cognitive performance (all P < 0.05). Notably, CSF CCL25 exhibited nonlinear relationships with Aβ and tau pathology, reaching a plateau as AD pathology increased. CSF CCL25 showed acceptable diagnostic accuracy in distinguishing amyloid-positive/negative (A ±) and tau-positive/negative (T ±) participants (area under the curve [AUC] = 0.71-0.77) and autopsy-confirmed AD cases (AUC = 0.77), with optimal performance in differentiating A + T + from A-T- participants (AUC = 0.82-0.85 with age and sex adjustment). Longitudinally, higher baseline CSF CCL25 predicted accelerated amyloid accumulation, hippocampal atrophy, and cognitive decline. Mediation analyses revealed that CCL25 partially mediated associations between Aβ pathology and tau pathology (mediating effect: 54.5%), neurodegeneration (18.2%), and cognitive decline (7.4%). Among 37 CSF CCL and CXCL chemokines examined, 28 were associated with at least one AD-related outcome, with CCL25 demonstrating the strongest associations overall. These findings suggest that CSF CCL25 is involved in early AD pathological progression and may serve as an inflammatory biomarker for diagnosis and monitoring of disease progression in AD.
Collapse
Affiliation(s)
- Yu-Han Chen
- Department of Human Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, 050017, China
- The First Clinical Medical School, Hebei North University, Zhangjiakou, 075000, China
| | - Zhi-Bo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Xi-Peng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Zhi-Qi Mao
- Department of Neurosurgery, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
32
|
Alanzi AR, Shahat AA, Alhaidhal BA, Aloatibi RM. Discovery of ROCK2 inhibitors through computational screening of ZINC database: Integrating pharmacophore modeling, molecular docking, and MD simulations. PLoS One 2025; 20:e0323781. [PMID: 40359277 PMCID: PMC12074392 DOI: 10.1371/journal.pone.0323781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Rho-associated protein kinase 2 (ROCK2) is a serine/threonine kinase that is crucial for regulating various physiological processes and is part of the Rho-associated coiled-coil kinase family. The dysregulation of ROCK2 has been associated with a range of diseases, making it a promising target for therapy. In this study, a chemical feature-based pharmacophore model was developed on the co-crystal ligand (5YS) of ROCK2 to conduct the virtual screening of ZINC database, resulting in 4809 hits that were further subjected to molecular docking to find the binding affinities with ROCK2 protein. The binding affinities of the hits were analyzed and compounds in the range of -11.55 to -9.91 kcal/mol were selected for further analysis. The ADMET analysis identified two promising compounds, whose binding stability with the ROCK2 protein was further evaluated using molecular dynamics (MD) simulations. Simulation results revealed that the selected compounds remained closely bound to protein indicating that they can act as lead compounds to control the biological activity of ROCK2. However, further in vitro investigation is required to test the biological efficacy of the reported compounds.
Collapse
Affiliation(s)
- Abdullah R. Alanzi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdelaaty A. Shahat
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | | |
Collapse
|
33
|
Mak E, Reid RI, Przybelski SA, Fought AM, Lesnick TG, Schwarz CG, Senjem ML, Raghavan S, Vemuri P, Jack CR, Min HK, Jain MK, Miyagawa T, Forsberg LK, Fields JA, Savica R, Graff-Radford J, Jones DT, Botha H, St Louis EK, Knopman DS, Ramanan VK, Dickson DW, Graff-Radford NR, Day GS, Ferman TJ, Petersen RC, Lowe VJ, Boeve BF, O'Brien JT, Kantarci K. Cortical microstructural abnormalities in dementia with Lewy bodies and their associations with Alzheimer's disease copathologies. NPJ Parkinsons Dis 2025; 11:124. [PMID: 40355490 PMCID: PMC12069582 DOI: 10.1038/s41531-025-00944-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 03/18/2025] [Indexed: 05/14/2025] Open
Abstract
Dementia with Lewy bodies (DLB) frequently coexists with Alzheimer's disease pathology, yet the pattern of cortical microstructural injury and its relationship with amyloid, tau, and cerebrovascular pathologies remains unclear. We applied neurite orientation dispersion and density imaging (NODDI) to assess cortical microstructural integrity in 57 individuals within the DLB spectrum and 57 age- and sex-matched cognitively unimpaired controls by quantifying mean diffusivity (MD), tissue-weighted neurite density index (tNDI), orientation dispersion index (ODI), and free water fraction (FWF). Amyloid and tau levels were measured using PiB and Flortaucipir PET imaging. Compared to controls, DLB exhibited increased MD and FWF, reduced tNDI across multiple regions, and focal ODI reductions in the occipital cortex. Structural equation modeling revealed that APOE genotype influenced amyloid levels, which elevated tau, leading to microstructural injury. These findings highlight the role of AD pathology in DLB neurodegeneration, advocating for multi-target therapeutic approaches addressing both AD and DLB-specific pathologies.
Collapse
Affiliation(s)
- Elijah Mak
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom.
| | - Robert I Reid
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Angela M Fought
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Hoon Ki Min
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Manoj K Jain
- Department of Radiology, Mayo Clinic, Jacksonville, FL, USA
| | - Toji Miyagawa
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Julie A Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Rodolfo Savica
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Erik K St Louis
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
- Center for Sleep Medicine, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Dennis W Dickson
- Laboratory of Medicine and Pathology, Mayo Clinic in Florida, Jacksonville, FL, USA
| | | | - Gregory S Day
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Tanis J Ferman
- Department of Psychiatry and Psychology, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Ronald C Petersen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
34
|
Zernov N, Melenteva D, Ghamaryan V, Makichyan A, Hunanyan L, Popugaeva E. N-N-Substituted Piperazine, Cmp2, Improves Cognitive and Motor Functions in 5xFAD Mice. Int J Mol Sci 2025; 26:4591. [PMID: 40429735 PMCID: PMC12111198 DOI: 10.3390/ijms26104591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 05/04/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
The piperazine derivative N-(2,6-difluorophenyl)-2-(4-phenylpiperazin-1-yl)propanamide (cmp2) has emerged as a potential transient receptor potential cation channel, subfamily C, member 6 (TRPC6) modulator, offering a promising pathway for Alzheimer's disease (AD) therapy. Our recent findings identify cmp2 as a novel compound with synaptoprotective effects in primary hippocampal cultures and effective blood-brain barrier (BBB) penetration. In vivo studies demonstrate that cmp2 (10 mg/kg, intraperitoneally) restores synaptic plasticity deficits in 5xFAD mice. This study further shows cmp2's selectivity towards tetrameric TRPC6 channel in silico. Acute administration of cmp2 is non-toxic, with no indications of chronic toxicity, and Ames testing confirms its lack of mutagenicity. Behavioral assays reveal that cmp2 improves cognitive functions in 5xFAD mice, including increased novel object recognition, better passing of the Morris water maze, and improved fear memory, as well as upregulation of motor function in beam walking tests. These findings suggest that cmp2 holds promise as a candidate for AD treatment.
Collapse
Affiliation(s)
- Nikita Zernov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| | - Daria Melenteva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| | - Viktor Ghamaryan
- Laboratory of Structural Bioinformatics, Institute of Biomedicine and Pharmacy, Russian-Armenian University, Yerevan 0051, Armenia (L.H.)
| | - Ani Makichyan
- Laboratory of Structural Bioinformatics, Institute of Biomedicine and Pharmacy, Russian-Armenian University, Yerevan 0051, Armenia (L.H.)
| | - Lernik Hunanyan
- Laboratory of Structural Bioinformatics, Institute of Biomedicine and Pharmacy, Russian-Armenian University, Yerevan 0051, Armenia (L.H.)
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| |
Collapse
|
35
|
Ding S, Choi SH, Miller YI. Amyloid β-Induced Inflammarafts in Alzheimer's Disease. Int J Mol Sci 2025; 26:4592. [PMID: 40429737 PMCID: PMC12111532 DOI: 10.3390/ijms26104592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
The formation of amyloid beta (Aβ) plaques is a central process in the development of Alzheimer's disease (AD). Although its causative role or the effectiveness of therapeutic targeting is still debated, the key involvement of Aβ in the pathogenesis of neuroinflammation and neurodegeneration in AD is broadly accepted. In this review, we emphasize the role of lipid rafts, both in APP cleavage producing Aβ in neurons and in mediating Aβ inflammatory signaling in microglia. We introduce the term inflammarafts to characterize the Aβ-driven formation of enlarged, cholesterol-rich lipid rafts in activated microglia, which support protein-protein and lipid-protein interactions of inflammatory receptors. Examples reviewed include toll-like receptors (TLR2, TLR4), scavenger receptors (CD36, RAGE), and TREM2. The downstream pathways lead to the production of cytokines and reactive oxygen species, intensifying neuroinflammation and resulting in neuronal injury and cognitive decline. We further summarize emerging therapeutic strategies and emphasize the utility of apolipoprotein A-I binding protein (AIBP) in selective targeting of inflammarafts and attenuation of microglia-driven inflammation. Unlike the targeting of a single inflammatory receptor or a secretase, selective disruption of inflammarafts and preservation of physiological lipid rafts offer a novel approach to targeting multiple components and processes that contribute to neuroinflammation in AD.
Collapse
Affiliation(s)
| | | | - Yury I. Miller
- Department of Medicine, University of California, San Diego, CA 92093, USA; (S.D.); (S.-H.C.)
| |
Collapse
|
36
|
Im D, Lee YE, Yoon G, Goddard WA, Choi TS, Kim HI. Antiparallel β-Sheet as a Key Motif of Amyloid-β Inhibitor Designed via Topological Peptide Reprogramming. Angew Chem Int Ed Engl 2025:e202504640. [PMID: 40345176 DOI: 10.1002/anie.202504640] [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: 02/26/2025] [Revised: 04/26/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Peptide inhibitor design targeting self-assembly of amyloid-β (Aβ) represents a promising strategy for suppressing the pathogenic mechanism of Alzheimer's disease (AD). Conventional approaches have primarily mimicked repetitive sequences found in fibrillar structures of Aβ aggregates. However, since the inherent flexibility of Aβ structures promotes the structural changes in the early-stage oligomerization, a structural modulation should be considered in the design of peptide inhibitors. Herein, we introduce topological reprogramming of peptides to control the structural transformation in pathogenic Aβ 1-42 (Aβ42). The eleven-residue peptide scaffold Pa11 (14HQKLVNFAEDV24) identified through the initial screening was dimerized via a disulfide bond. The dimerization stabilizes Aβ42 into higher order structures by promoting antiparallel β-sheet conformations, thereby significantly suppressing Aβ42 aggregation. Our approach underscores that modification in peptide connectivity would be a breakthrough for controlling the intrinsic flexibility of Aβ, surpassing the limitation in conventional, one-dimensional peptide building block.
Collapse
Affiliation(s)
- Dongjoon Im
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
- Division of Chemistry and Chemical Engineering and Materials Process and Simulation Center, California Institute of Technology, Pasadena, CA 91125, USA
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Ye Eun Lee
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Gyusub Yoon
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - William A Goddard
- Division of Chemistry and Chemical Engineering and Materials Process and Simulation Center, California Institute of Technology, Pasadena, CA 91125, USA
| | - Tae Su Choi
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
- Center for Proteogenome Research, Korea University, Seoul, 02841, Republic of Korea
- Center for Mechanogenome Research, Korea University, Seoul, 02841, Republic of Korea
| | - Hugh I Kim
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
- Division of Chemistry and Chemical Engineering and Materials Process and Simulation Center, California Institute of Technology, Pasadena, CA 91125, USA
- Center for Proteogenome Research, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
37
|
Alanzi AR, Alhaidhal BA, Aloatibi RM. Identification of SIRT3 modulating compounds in deep-sea fungi metabolites: Insights from molecular docking and MD simulations. PLoS One 2025; 20:e0323107. [PMID: 40338931 PMCID: PMC12061134 DOI: 10.1371/journal.pone.0323107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/02/2025] [Indexed: 05/10/2025] Open
Abstract
SIRT3, a crucial deacetylase that plays a key role in regulating mitochondrial acetylation, is tightly linked to metabolic processes and is essential for the maintenance of eukaryotic life. SIRT3 is a potential therapeutic target due to its key role in various diseases, including ageing, heart disease, cancer, and metabolic disorders. In this work, we aimed to identify potential SIRT3 inhibitors from the deep-sea fungal metabolites by employing molecular docking and ADMET analysis. Based on the binding affinities, ten compounds were selected whose docking scores were in the range of -9.693 to -8.327 kcal/mol. Further, four compounds Penipanoid C, Penicillactam, Quinolonimide, and Brevianamide R were selected based on the ADMET properties and subjected to Molecular dynamics simulations to assess the stability of these molecules with target. The stability analysis indicated that the selected compounds could act as lead compounds during in vitro assays to advance these drug candidates towards clinical drug development.
Collapse
Affiliation(s)
- Abdullah R. Alanzi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | | |
Collapse
|
38
|
Ortiz AA, Murtishaw AS, Beckholt M, Salazar AM, Osse AML, Kinney JW. Impact of chronic hyperglycemia and high-fat diet on Alzheimer's disease-related pathology in CX3CR1 knockout mice. Metab Brain Dis 2025; 40:197. [PMID: 40332622 DOI: 10.1007/s11011-025-01618-6] [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: 05/05/2024] [Accepted: 04/18/2025] [Indexed: 05/08/2025]
Abstract
Diabetes mellitus (DM), obesity, and metabolic syndrome are related disorders with wide-ranging and devastating effects that are comorbid with many other diseases. Clinical and epidemiological studies have found that type II diabetes mellitus (T2DM), including chronic hyperglycemia and hyperinsulinemia, significantly increases the risk of Alzheimer's disease (AD) and other forms of dementia in the elderly. Insulin has slightly different functions in the peripheral body than in the central nervous system and the dysregulation of these functions may contribute to the onset and progression of late-life neurodegenerative disease. To investigate cognitive function and AD-related disease pathology, we utilized two different models of key features of diabetes, one model characterized by hyperglycemia resulting from a diabetogenic compound that selectively targets insulin-producing pancreatic β-cells, and the other model based on diet-induced obesity. Additionally, these diabetic models were combined with fractalkine receptor knockout mice (CX3CR1-/-), a genetic mouse model of inflammation, to explore the additive effects of multiple AD risk factors. The CX3CR1 receptor has been implicated in modulating neuroinflammation associated with AD, and its dysregulation can exacerbate metabolic disturbances and neurodegenerative markers. We found that diabetic-status, regardless of whether it was drug- or diet-induced, resulted in profound impairments in learning and memory and AD-related alterations within the hippocampus.
Collapse
Affiliation(s)
- Andrew Adonay Ortiz
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA.
| | - Andrew Scott Murtishaw
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Monica Beckholt
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Arnold Maloles Salazar
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Amanda Marie Leisgang Osse
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Jefferson William Kinney
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA
| |
Collapse
|
39
|
Catto F, Kirschenbaum D, Economides AE, Reuss AM, Trevisan C, Caredio D, Dadgar-Kiani E, Mirzet D, Frick L, Weber-Stadlbauer U, Litvinov S, Koumoutsakos P, Lee JH, Aguzzi A. Quantitative 3D histochemistry reveals region-specific amyloid-β reduction by the antidiabetic drug netoglitazone. PLoS One 2025; 20:e0309489. [PMID: 40327707 PMCID: PMC12054868 DOI: 10.1371/journal.pone.0309489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/29/2025] [Indexed: 05/08/2025] Open
Abstract
A hallmark of Alzheimer's disease (AD) is the extracellular aggregation of toxic amyloid-beta (Aβ) peptides in form of plaques. Here, we identify netoglitazone, an antidiabetic compound previously tested in humans, as an Aβ aggregation antagonist. Netoglitazone improved cognition and reduced microglia activity in a mouse model of AD. Using quantitative whole-brain three-dimensional histology (Q3D), we precisely identified brain regions where netoglitazone reduced the number and size of Aβ plaques. We demonstrate the utility of Q3D in preclinical drug evaluation for AD by providing a high-resolution brain-wide view of drug efficacy. Applying Q3D has the potential to improve pre-clinical drug evaluation by providing information that can help identify mechanisms leading to brain region-specific drug efficacy.
Collapse
Affiliation(s)
- Francesca Catto
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- IMAI MedTech GmbH, Zurich, Switzerland
| | - Daniel Kirschenbaum
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Athena E. Economides
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anna Maria Reuss
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Chiara Trevisan
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Davide Caredio
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ehsan Dadgar-Kiani
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Delic Mirzet
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lukas Frick
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Sergey Litvinov
- Computational Science and Engineering Laboratory, School of Engineering and Applied Sciences, Harvard University, Cambridge, United States of America
| | - Petros Koumoutsakos
- Computational Science and Engineering Laboratory, School of Engineering and Applied Sciences, Harvard University, Cambridge, United States of America
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, California, United States of America
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
- Department of Electrical Engineering, Stanford University, Stanford, California, United States of America
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
40
|
Zhao C, Yue J, Xie Y, Liu B, Xu S, Zhi D, Wang D. A Ginsenoside Composition Ameliorated Aβ and Tau Aggregation via Autophagy Lysosome Pathway. Mol Neurobiol 2025:10.1007/s12035-025-05017-x. [PMID: 40327308 DOI: 10.1007/s12035-025-05017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by the abnormal deposition of amyloid-beta (Aβ) peptides and neurofibrillary tangles (NFTs). Ginsenosides, the primary active constituents in ginseng, exhibit potential in combating AD. In our previous work, the ginsenoside SumI was demonstrated to have superior anti-AD activity compared to other ginsenosides when used alone. This study revealed that SumI effectively decreased the lysosomal pH, promoted autophagosome formation, increased autophagic flux, and facilitated the transport of misfolded proteins to lysosomes for degradation in Caenorhabditis elegans. SumI activated the HLH-30 transcription factor by triggering a lipid-catabolic response akin to starvation. bec-1 RNAi significantly abrogated the anti-AD effect of SumI. Our findings indicate that SumI mitigated protein aggregation by activating the autophagy-lysosome pathway in C. elegans and provide scientific evidence that ginsenoside composition could be a potential therapeutic agent for treating or preventing AD.
Collapse
Affiliation(s)
- Chengmu Zhao
- School of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu, 730000, China
- Frontiers Science Center for Rare Isotopes, No. 222 Tianshui South Road, Lanzhou, Gansu, 730000, China
| | - Juan Yue
- School of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu, 730000, China
| | - Yu Xie
- School of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu, 730000, China
- Frontiers Science Center for Rare Isotopes, No. 222 Tianshui South Road, Lanzhou, Gansu, 730000, China
| | - Bo Liu
- School of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu, 730000, China
- Frontiers Science Center for Rare Isotopes, No. 222 Tianshui South Road, Lanzhou, Gansu, 730000, China
| | - Shuaishuai Xu
- School of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu, 730000, China
| | - Dejuan Zhi
- School of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu, 730000, China.
- Frontiers Science Center for Rare Isotopes, No. 222 Tianshui South Road, Lanzhou, Gansu, 730000, China.
| | - Dongsheng Wang
- School of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu, 730000, China.
- Frontiers Science Center for Rare Isotopes, No. 222 Tianshui South Road, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
41
|
Sandberg A, Puttagunta S, Duval N, Fleming H, Koza L, Hieber K, Holsopple J, Reyna M, Paredes D, Linseman DA. Immunocal ®, a cysteine-rich whey protein, rescues reelin and reduces amyloid plaque burden in a transgenic amyloid-β protein precursor (hAβPP SweInd) mouse model of Alzheimer's disease. J Alzheimers Dis 2025:13872877251338323. [PMID: 40325994 DOI: 10.1177/13872877251338323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
BackgroundDeficits in Reelin expression play a significant role in the pathogenesis of various neurological disorders, including schizophrenia and Alzheimer's disease (AD). Notably, Reelin-expressing neurons of the entorhinal cortex layer II are among the first to be affected in AD.ObjectiveStrategies aimed at correcting deficits in Reelin might provide a novel therapeutic approach for AD.MethodsHere, we examined the effects of the whey protein supplement and glutathione (GSH) precursor, Immunocal®, on Reelin expression both in vitro in hippocampal-entorhinal cortex slices from rat brain and in vivo in the hAβPPSweInd (J20) mouse model of AD.ResultsIncubation of brain slices with Immunocal® increased Reelin expression at the mRNA and protein levels. Oral treatment with Immunocal®, given ad libitum in drinking water beginning at 3 months of age, corrected a deficit in cortical GSH levels observed in untreated mice and preserved Reelin expression in the hippocampal-entorhinal cortex sub-region of 5-month-old J20 mice. We also assessed the long-term effects of Immunocal® by treating J20 mice from 3 months old to 12 months old. Long-term Immunocal® treatment preserved brain GSH and rescued Reelin mRNA and protein expression, while significantly reducing amyloid plaque formation in the entorhinal cortex and hippocampus of AD mice.ConclusionsThese findings suggest that Immunocal® promotes Reelin expression in vitro and sustains brain GSH and Reelin expression while diminishing amyloid plaque load in the entorhinal cortex and hippocampus of J20 mice. Thus, Immunocal® offers a promising therapeutic approach to enhance Reelin expression and curtail amyloid deposition in AD.
Collapse
Affiliation(s)
- Alexandra Sandberg
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | | | - Nathan Duval
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Holly Fleming
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Lilia Koza
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Kade Hieber
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Jessica Holsopple
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Michael Reyna
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Daniel Paredes
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Daniel A Linseman
- Department of Biological Sciences, University of Denver, Denver, CO, USA
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| |
Collapse
|
42
|
Lozupone M, Dibello V, Resta E, Sardone R, Castellana F, Zupo R, Lampignano L, Bortone I, Mollica A, Berardino G, Altamura M, Bellomo A, Daniele A, Solfrizzi V, Panza F. Uncertainties in anti-amyloid monoclonal antibody therapy for Alzheimer's disease: the challenges ahead. Expert Rev Neurother 2025:1-11. [PMID: 40314575 DOI: 10.1080/14737175.2025.2500752] [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: 02/09/2025] [Revised: 04/04/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD), the leading cause of dementia, poses a significant burden on patients, caregivers, and healthcare systems worldwide. After two decades of extensive efforts, we are still without significantly effective disease-modifying drugs for AD. Although brain amyloid-β (Aβ) accumulation may predict cognitive decline, several drug candidates, including anti-Aβ monoclonal antibodies, have been developed and tested to reduce Aβ plaque burden effective, but without significant clinical success. AREAS COVERED The following review presents and discusses anti-Aβ monoclonal antibody therapeutics used to treat AD. The article considers both current approaches and alternatives. This article is multiple database searches (MEDLINE, EMBASE, Scopus, Ovid and Google Scholar) on all the available literature up to 1 February 2025. EXPERT OPINION Randomized clinical trials (RCTs) of anti-Aβ drugs in AD have not fully validated the Aβ cascade hypothesis. Nevertheless, eight anti-Aβ monoclonal antibodies have, thus far, made it to Phase III RCTs. Moving forward, the use of the Apolipoprotein E genotype and tau protein as alternative biomarkers can assist clinicians in providing patients with even more individualized and efficacious anti-Aβ monoclonal antibodies dosing regimens and reduce the risk of serious amyloid-related imaging abnormalities.
Collapse
Affiliation(s)
- Madia Lozupone
- Department of Translational Biomedicine and Neuroscience "DiBrain", University of Bari Aldo Moro, Bari, Italy
| | - Vittorio Dibello
- Department of Interdisciplinary Medicine (DIM), "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Emanuela Resta
- Translational Medicine and Health System Management, Department of Economy, University of Foggia, Foggia, Italy
| | - Rodolfo Sardone
- Unit of Statistics and Epidemiology, Local Healthcare Authority of Taranto, Taranto, Italy
| | - Fabio Castellana
- Department of Interdisciplinary Medicine (DIM), "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
| | - Roberta Zupo
- Department of Interdisciplinary Medicine (DIM), "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
| | - Luisa Lampignano
- Unit of Statistics and Epidemiology, Local Healthcare Authority of Taranto, Taranto, Italy
| | - Ilaria Bortone
- Local Healthcare Authority of Bari, ASL Bari, Bari, Italy
| | - Anita Mollica
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giuseppe Berardino
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Mario Altamura
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonio Daniele
- Department of Neuroscience, Catholic University of Sacred Heart, Rome, Italy
- Neurology Unit, IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Vincenzo Solfrizzi
- Department of Interdisciplinary Medicine (DIM), "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Panza
- Department of Interdisciplinary Medicine (DIM), "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
43
|
Jeong H, Pan Y, Akhter F, Volkow ND, Zhu D, Du C. Evidence of cortical vascular impairments in early stage of Alzheimer's transgenic mice: Optical imaging. J Cereb Blood Flow Metab 2025; 45:960-976. [PMID: 39696904 PMCID: PMC12035375 DOI: 10.1177/0271678x241304893] [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: 12/21/2023] [Revised: 08/20/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024]
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder with progressive cognitive decline, remains clinically challenging with limited understanding of etiology and interventions. Clinical studies have reported vascular defects prior to other pathological manifestations of AD, leading to the "Vascular Hypothesis" for the disorder. However, in vivo assessments of cerebral vasculature in AD rodent models have been constrained by limited spatiotemporal resolution or field of view of conventional imaging. We herein employed two in vivo imaging technologies, Dual-Wavelength Imaging and Optical Coherence Doppler Tomography, to evaluate cerebrovascular reactivity (CVR) to vasoconstrictive cocaine and vasodilatory hypercapnia challenges and to detect resting 3D cerebral blood flow (CBF) in living transgenic AD mice at capillary resolution. Results showed that CVR to cocaine and hypercapnia was significantly attenuated in 7-10 months old AD mice vs controls, indicating reduced vascular flexibility and reactivity. Additionally, in the AD mice, arterial CBF velocities were slower and the microvascular density in cortex was decreased compared to controls. These results reveal significant vascular impairments including reduced CVR and resting CBF in early-staged AD mice. Hence, this cutting-edge in vivo optical imaging offers an innovative venue for detecting early neurovascular dysfunction in AD brain with translational potential.
Collapse
Affiliation(s)
- Hyomin Jeong
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Yingtian Pan
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Firoz Akhter
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Donghui Zhu
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Congwu Du
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| |
Collapse
|
44
|
Kelliny S, Zhou X, Bobrovskaya L. Alzheimer's Disease and Frontotemporal Dementia: A Review of Pathophysiology and Therapeutic Approaches. J Neurosci Res 2025; 103:e70046. [PMID: 40387258 PMCID: PMC12087441 DOI: 10.1002/jnr.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/01/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
Alzheimer's disease (AD) is a devastating form of dementia, with the number of affected individuals rising sharply. The main hallmarks of the disease include amyloid-beta plaque deposits and neurofibrillary tangles consisting of hyperphosphorylated tau protein, besides other pathological features that contribute to the disease's complexity. The causes of sporadic AD are multifactorial and mostly age-related and involve risk factors such as diabetes and cardiovascular or cerebrovascular disorders. Frontotemporal dementia (FTD) is another type of dementia characterized by a spectrum of behaviors, memory, and motor abnormalities and associated with abnormal depositions of protein aggregation, including tau protein. Currently approved medications are symptomatic, and no disease-modifying therapy is available to halt the disease progression. Therefore, the development of multi-targeted therapeutic approaches could hold promise for the treatment of AD and other neurodegenerative disorders, including tauopathies. In this article, we will discuss the pathophysiology of AD and FTD, the proposed hypotheses, and current therapeutic approaches, highlighting the development of novel drug candidates and the progress of clinical trials in this field of research.
Collapse
Affiliation(s)
- Sally Kelliny
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- Faculty of PharmacyAssiut UniversityAssiutEgypt
| | - Xin‐Fu Zhou
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
45
|
Shen Y, Fan N, Ma S, Cheng X, Yang X, Wang G. Gut Microbiota Dysbiosis: Pathogenesis, Diseases, Prevention, and Therapy. MedComm (Beijing) 2025; 6:e70168. [PMID: 40255918 PMCID: PMC12006732 DOI: 10.1002/mco2.70168] [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: 08/04/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/22/2025] Open
Abstract
Dysbiosis refers to the disruption of the gut microbiota balance and is the pathological basis of various diseases. The main pathogenic mechanisms include impaired intestinal mucosal barrier function, inflammation activation, immune dysregulation, and metabolic abnormalities. These mechanisms involve dysfunctions in the gut-brain axis, gut-liver axis, and others to cause broader effects. Although the association between diseases caused by dysbiosis has been extensively studied, many questions remain regarding the specific pathogenic mechanisms and treatment strategies. This review begins by examining the causes of gut microbiota dysbiosis and summarizes the potential mechanisms of representative diseases caused by microbiota imbalance. It integrates clinical evidence to explore preventive and therapeutic strategies targeting gut microbiota dysregulation, emphasizing the importance of understanding gut microbiota dysbiosis. Finally, we summarized the development of artificial intelligence (AI) in the gut microbiota research and suggested that it will play a critical role in future studies on gut dysbiosis. The research combining multiomics technologies and AI will further uncover the complex mechanisms of gut microbiota dysbiosis. It will drive the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Yao Shen
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
| | - Nairui Fan
- Basic Medical College of Jiamusi UniversityHeilongjiangChina
| | - Shu‐xia Ma
- Basic Medical College of Jiamusi UniversityHeilongjiangChina
| | - Xin Cheng
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
- International SchoolGuangzhou Huali College, ZengchengGuangzhouChina
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryGuangdong Second Provincial General HospitalSchool of MedicineJinan UniversityGuangzhouChina
| |
Collapse
|
46
|
Salloway S, Pain A, Lee E, Papka M, Ferguson MB, Wang H, Hu H, Lu M, Oru E, Ardayfio PA, Hoban DB, Collins EC, Brooks DA, Sims JR. TRAILBLAZER-ALZ 4: A phase 3 trial comparing donanemab with aducanumab on amyloid plaque clearance in early, symptomatic Alzheimer's disease. Alzheimers Dement 2025; 21:e70293. [PMID: 40390253 PMCID: PMC12089073 DOI: 10.1002/alz.70293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/21/2025]
Abstract
INTRODUCTION The phase 3, open-label TRAILBLAZER-ALZ 4 study compared the effect of donanemab versus aducanumab on amyloid plaque (AP) clearance in participants with early symptomatic Alzheimer's disease. METHODS Participants (n = 148) were randomized 1:1 to receive intravenous donanemab (700 mg every 4 weeks for three doses, then 1400 mg every 4 weeks thereafter) or aducanumab (per label). AP was measured with florbetapir F 18 positron emission tomography. AP clearance was defined as < 24.1 Centiloids. RESULTS At 6, 12, and 18 months, AP clearance was achieved in 37.9%, 70.0%, and 76.8%, respectively, of donanemab-treated participants versus 1.6%, 24.6%, and 43.1% of aducanumab-treated participants (P < 0.001). Median time to clearance was 359 versus 568 days for donanemab- versus aducanumab-treated participants (P < 0.001). Amyloid-related imaging abnormality (ARIA)-edema/effusion occurred in 23.9% and 34.8% of donanemab- and aducanumab-treated participants, respectively. DISCUSSION Donanemab treatment resulted in earlier and greater AP clearance compared to aducanumab. ARIA frequencies were consistent with prior studies. CLINICAL TRIAL REGISTRATION No: NCT05108922, TRAILBLAZER-ALZ 4 HIGHLIGHTS: Here we report the first direct comparator study of two amyloid-targeting therapies. This was the first investigation of donanemab on biomarker efficacy regardless of tau levels. Donanemab demonstrated superiority over aducanumab in amyloid plaque (AP) clearance. The depth and speed of AP removal did not affect amyloid-related imaging abnormality risk or incidence.
Collapse
Affiliation(s)
- Stephen Salloway
- Department of Neurology and Department of PsychiatryAlpert Medical School of Brown UniversityProvidenceRhode IslandUSA
- Butler HospitalProvidenceRhode IslandUSA
| | | | - Elly Lee
- Irvine Clinical ResearchIrvineCaliforniaUSA
| | - Michelle Papka
- The Cognitive and Research Center of New Jersey LLCSpringfieldNew JerseyUSA
| | | | - Hong Wang
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | - Haoyan Hu
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | - Ming Lu
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | - Ena Oru
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | | | | | | | | | | |
Collapse
|
47
|
Castro-Aldrete L, Einsiedler M, Novakova Martinkova J, Depypere H, Alvin Ang TF, Mielke MM, Sindi S, Eyre HA, Au R, Schumacher Dimech AM, Dé A, Szoeke C, Tartaglia MC, Santuccione Chadha A. Alzheimer disease seen through the lens of sex and gender. Nat Rev Neurol 2025; 21:235-249. [PMID: 40229578 DOI: 10.1038/s41582-025-01071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2025] [Indexed: 04/16/2025]
Abstract
Alzheimer disease (AD) is a life-limiting neurodegenerative disorder that disproportionately affects women. Indeed, sex and gender are emerging as crucial modifiers of diagnostic and therapeutic pathways in AD. This Review provides an overview of the interactions of sex and gender with important developments in AD and offers insights into priorities for future research to facilitate the development and implementation of personalized approaches in the shifting paradigm of AD care. In particular, this Review focuses on the influence of sex and gender on important advances in the treatment and diagnosis of AD, including disease-modifying therapies, fluid-based biomarkers, cognitive assessment tools and multidomain lifestyle interventional studies.
Collapse
Affiliation(s)
| | | | - Julie Novakova Martinkova
- Women's Brain Foundation, Basel, Switzerland
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic
| | - Herman Depypere
- Department of Gynecology, Breast and Menopause Clinic, University Hospital, Coupure Menopause Centre, Ghent, Belgium
| | - Ting Fang Alvin Ang
- Department of Anatomy and Neurobiology and Slone Center of Epidemiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Shireen Sindi
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- The Ageing Epidemiology Research Unit, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Harris A Eyre
- Neuro-Policy Program, Center for Health and Biosciences, The Baker Institute for Public Policy, Rice University, Houston, TX, USA
- Euro-Mediterranean Economists Association, Barcelona, Spain
| | - Rhoda Au
- Department of Anatomy and Neurobiology, Neurology, Medicine and Epidemiology, Boston University Chobanian and Avedisian School of Medicine and School of Public Health, Boston, MA, USA
| | - Anne Marie Schumacher Dimech
- Women's Brain Foundation, Basel, Switzerland
- Faculty of Health Sciences and Medicine, University of Lucerne, Lucerne, Switzerland
| | - Anna Dé
- Women's Brain Foundation, Basel, Switzerland
| | | | - Maria Carmela Tartaglia
- Women's Brain Foundation, Basel, Switzerland
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
48
|
Noguchi‐Shinohara M, Shuta K, Murakami H, Mori Y, Komatsu J, Kobayashi C, Hersch S, Horie K, Ono K. Lecanemab-Associated Amyloid-β Protofibril in Cerebrospinal Fluid Correlates with Biomarkers of Neurodegeneration in Alzheimer's Disease. Ann Neurol 2025; 97:993-1006. [PMID: 39761671 PMCID: PMC12010060 DOI: 10.1002/ana.27175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 04/22/2025]
Abstract
OBJECTIVE The Clarity AD phase III trial showed that lecanemab reduced amyloid markers in early Alzheimer's disease (AD) and resulted in less decline on measures of cognition and function than placebo. Herein, we aimed to characterize amyloid-β (Aβ) protofibril (PF) captured by lecanemab in human cerebrospinal fluid (CSF) from living participants with different stages in AD, which enable an enhanced understanding of the dynamic changes of lecanemab-associated Aβ-PF (Lec-PF) in vivo. METHODS We newly developed a unique and highly sensitive immunoassay method using lecanemab that selectively captures Lec-PF. The CSF level of Lec-PF, Aβ42, Aβ40, p-tau181, p-tau 217, total tau, and neurogranin were measured in Japanese participants (n = 163). The participants in this study consisted of 48 cognitively unimpaired Aβ-negative (CU-), 8 cognitively impaired diagnosed as suspected non-Alzheimer's disease pathophysiology, 9 cognitively unimpaired Aβ-positive (CU+), 34 Aβ-positive with mild cognitive impairment (MCI+), and 64 Aβ-positive with AD dementia (AD+). RESULTS The CSF Lec-PF levels significantly increased in the groups of MCI+ and AD+ compared with CU- group. Notably, CSF Lec-PF showed modest correlation with plaque-associated biomarkers in Aβ-positive participants and stronger correlation with neurodegeneration biomarkers, such as CSF total tau and neurogranin, suggesting that CSF Lec-PF levels proximally reflect neurodegeneration as well as the amount of senile amyloid plaques. INTERPRETATION This is the first report describing Aβ-PF species captured by lecanemab in human CSF and supporting that Lec-PF is correlated with neurodegeneration in AD and may explain the mechanism of the clinical effect of lecanemab. ANN NEUROL 2025;97:993-1006.
Collapse
Affiliation(s)
| | - Kazuyoshi Shuta
- Deep Human Biology Learning (DHBL)Eisai Co., LtdTsukubaJapan
| | - Hidetomo Murakami
- Department of Neurology, School of MedicineShowa UniversityTokyoJapan
| | - Yukiko Mori
- Department of Neurology, School of MedicineShowa UniversityTokyoJapan
| | - Junji Komatsu
- Department of NeurologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| | | | - Steven Hersch
- Deep Human Biology Learning (DHBL)Eisai Inc.NutleyNJUSA
| | - Kanta Horie
- Deep Human Biology Learning (DHBL)Eisai Inc.NutleyNJUSA
| | - Kenjiro Ono
- Department of NeurologyKanazawa University Graduate School of Medical SciencesKanazawaJapan
| |
Collapse
|
49
|
Yang X, Yao K, Zhang M, Zhang W, Zu H. New insight into the role of altered brain cholesterol metabolism in the pathogenesis of AD: A unifying cholesterol hypothesis and new therapeutic approach for AD. Brain Res Bull 2025; 224:111321. [PMID: 40164234 DOI: 10.1016/j.brainresbull.2025.111321] [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: 01/30/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
The dysregulation of cholesterol metabolism homeostasis has been universally suggested in the aeotiology of Alzheimer's disease (AD). Initially, studies indicate that alteration of serum cholesterol level might contribute to AD. However, because blood-brain barrier impedes entry of plasma cholesterol, brain cells are not directly influenced by plasma cholesterol. Furthermore, mounting evidences suggest a link between alteration of brain cholesterol metabolism and AD. Interestingly, Amyloid-β proteins (Aβ) can markedly inhibit cellular cholesterol biosynthesis and lower cellular cholesterol content in cultured cells. And Aβ overproduction/overload induces a significant decrease of brain cellular cholesterol content in familial AD (FAD) animals. Importantly, mutations or polymorphisms of genes related to brain cholesterol transportation, such as ApoE4, ATP binding cassette (ABC) transporters, low-density lipoprotein receptor (LDLR) family and Niemann-Pick C disease 1 or 2 (NPC1/2), obviously lead to decreased brain cholesterol transport, resulting in brain cellular cholesterol loss, which could be tightly associated with AD pathological impairments. Additionally, accumulating data show that there are reduction of brain cholesterol biosynthesis and/or disorder of brain cholesterol trafficking in a variety of sporadic AD (SAD) animals and patients. Collectively, compelling evidences indicate that FAD and SAD could share one common and overlapping neurochemical mechanism: brain neuronal/cellular cholesterol deficiency. Therefore, accumulated evidences strongly support a novel hypothesis that deficiency of brain cholesterol contributes to the onset and progression of AD. This review highlights the pivotal role of brain cholesterol deficiency in the pathogenesis of AD. The hypothesis offers valuable insights for the future development of AD treatment.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China; Department of Neurology, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200237, China
| | - Kai Yao
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Mengqi Zhang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Wenbin Zhang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Hengbing Zu
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China.
| |
Collapse
|
50
|
Chen YH, Wang ZB, Liu XP, Mao ZQ. Cerebrospinal fluid LMO4 as a synaptic biomarker linked to Alzheimer's disease pathology and cognitive decline. J Alzheimers Dis 2025; 105:216-227. [PMID: 40105503 DOI: 10.1177/13872877251326286] [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: 03/20/2025]
Abstract
BackgroundLIM-domain-only 4 (LMO4) is involved in neurodevelopment and synaptic plasticity, but its role in the pathogenesis of Alzheimer's disease (AD) remains unclear.ObjectiveTo investigate the association between cerebrospinal fluid (CSF) LMO4 levels and core AD biomarkers, neurodegeneration, and cognitive decline.MethodsWe included 703 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Associations between CSF LMO4 and AD biomarkers (Aβ42, Ptau181, amyloid PET) and postmortem neuropathology were evaluated. We also explored cross-sectional and longitudinal associations between CSF LMO4 and neurodegeneration and cognitive function. Receiver operating characteristic (ROC) analysis assessed the diagnostic accuracy of CSF LMO4 in distinguishing Aβ-positive from Aβ-negative participants and amyloid PET-confirmed AD cases. Mediation analysis explored the potential mediating role of CSF LMO4 between Aβ pathology and tau pathology.ResultsLMO4 levels were decreased in participants with abnormal Aβ levels and cognitive impairment. Lower CSF LMO4 levels were associated with increased Aβ and tau pathology, brain atrophy, cognitive decline, and postmortem neuropathology. CSF LMO4 partially mediated the relationship between Aβ and tau pathology and demonstrated acceptable discriminative ability in distinguishing Aβ-positive from Aβ-negative participants and amyloid PET-confirmed AD from non-AD cases.ConclusionsCSF LMO4 plays a crucial role in the pathogenesis and progression of AD and may represent a potential therapeutic target for AD treatment.
Collapse
Affiliation(s)
- Yu-Han Chen
- The First Clinical Medical School, Hebei North University, Zhangjiakou, China
| | - Zhi-Bo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Xi-Peng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Hebei North University, Hebei, Zhangjiakou, China
| | - Zhi-Qi Mao
- Department of Neurosurgery, the First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|