1
|
Spicer MM, Yang J, Fu D, DeVore AN, Lauffer M, Atasoy NS, Atasoy D, Fisher RA. Regulator of G protein signaling 6 mediates exercise-induced recovery of hippocampal neurogenesis, learning, and memory in a mouse model of Alzheimer's disease. Neural Regen Res 2025; 20:2969-2981. [PMID: 39248184 PMCID: PMC11826473 DOI: 10.4103/nrr.nrr-d-23-01993] [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: 12/05/2023] [Revised: 07/16/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202510000-00027/figure1/v/2024-11-26T163120Z/r/image-tiff Hippocampal neuronal loss causes cognitive dysfunction in Alzheimer's disease. Adult hippocampal neurogenesis is reduced in patients with Alzheimer's disease. Exercise stimulates adult hippocampal neurogenesis in rodents and improves memory and slows cognitive decline in patients with Alzheimer's disease. However, the molecular pathways for exercise-induced adult hippocampal neurogenesis and improved cognition in Alzheimer's disease are poorly understood. Recently, regulator of G protein signaling 6 (RGS6) was identified as the mediator of voluntary running-induced adult hippocampal neurogenesis in mice. Here, we generated novel RGS6 fl/fl ; APP SWE mice and used retroviral approaches to examine the impact of RGS6 deletion from dentate gyrus neuronal progenitor cells on voluntary running-induced adult hippocampal neurogenesis and cognition in an amyloid-based Alzheimer's disease mouse model. We found that voluntary running in APP SWE mice restored their hippocampal cognitive impairments to that of control mice. This cognitive rescue was abolished by RGS6 deletion in dentate gyrus neuronal progenitor cells, which also abolished running-mediated increases in adult hippocampal neurogenesis. Adult hippocampal neurogenesis was reduced in sedentary APP SWE mice versus control mice, with basal adult hippocampal neurogenesis reduced by RGS6 deletion in dentate gyrus neural precursor cells. RGS6 was expressed in neurons within the dentate gyrus of patients with Alzheimer's disease with significant loss of these RGS6-expressing neurons. Thus, RGS6 mediated voluntary running-induced rescue of impaired cognition and adult hippocampal neurogenesis in APP SWE mice, identifying RGS6 in dentate gyrus neural precursor cells as a possible therapeutic target in Alzheimer's disease.
Collapse
Affiliation(s)
- Mackenzie M. Spicer
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Molecular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jianqi Yang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Daniel Fu
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Alison N. DeVore
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Marisol Lauffer
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Neural Circuits and Behavior Core, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Nilufer S. Atasoy
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Rory A. Fisher
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
2
|
Zhang Y, Liang Y, Gu Y. The dopaminergic system and Alzheimer's disease. Neural Regen Res 2025; 20:2495-2512. [PMID: 39314145 PMCID: PMC11801300 DOI: 10.4103/nrr.nrr-d-24-00230] [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: 02/25/2024] [Revised: 06/21/2024] [Accepted: 07/31/2024] [Indexed: 09/25/2024] Open
Abstract
Alzheimer's disease is a common neurodegenerative disorder in older adults. Despite its prevalence, its pathogenesis remains unclear. In addition to the most widely accepted causes, which include excessive amyloid-beta aggregation, tau hyperphosphorylation, and deficiency of the neurotransmitter acetylcholine, numerous studies have shown that the dopaminergic system is also closely associated with the occurrence and development of this condition. Dopamine is a crucial catecholaminergic neurotransmitter in the human body. Dopamine-associated treatments, such as drugs that target dopamine receptor D and dopamine analogs, can improve cognitive function and alleviate psychiatric symptoms as well as ameliorate other clinical manifestations. However, therapeutics targeting the dopaminergic system are associated with various adverse reactions, such as addiction and exacerbation of cognitive impairment. This review summarizes the role of the dopaminergic system in the pathology of Alzheimer's disease, focusing on currently available dopamine-based therapies for this disorder and the common side effects associated with dopamine-related drugs. The aim of this review is to provide insights into the potential connections between the dopaminergic system and Alzheimer's disease, thus helping to clarify the mechanisms underlying the condition and exploring more effective therapeutic options.
Collapse
Affiliation(s)
- Yuhan Zhang
- International Medical College, Chongqing Medical University, Chongqing, China
| | - Yuan Liang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yixue Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
3
|
Long Z, Ge C, Zhao Y, Liu Y, Zeng Q, Tang Q, Dong Z, He G. Enhanced autophagic clearance of amyloid-β via histone deacetylase 6-mediated V-ATPase assembly and lysosomal acidification protects against Alzheimer's disease in vitro and in vivo. Neural Regen Res 2025; 20:2633-2644. [PMID: 38993141 PMCID: PMC11801289 DOI: 10.4103/nrr.nrr-d-23-01633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/05/2024] [Accepted: 03/29/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202509000-00025/figure1/v/2024-11-05T132919Z/r/image-tiff Recent studies have suggested that abnormal acidification of lysosomes induces autophagic accumulation of amyloid-β in neurons, which is a key step in senile plaque formation. Therefore, restoring normal lysosomal function and rebalancing lysosomal acidification in neurons in the brain may be a new treatment strategy for Alzheimer's disease. Microtubule acetylation/deacetylation plays a central role in lysosomal acidification. Here, we show that inhibiting the classic microtubule deacetylase histone deacetylase 6 with an histone deacetylase 6 shRNA or thehistone deacetylase 6 inhibitor valproic acid promoted lysosomal reacidification by modulating V-ATPase assembly in Alzheimer's disease. Furthermore, we found that treatment with valproic acid markedly enhanced autophagy, promoted clearance of amyloid-β aggregates, and ameliorated cognitive deficits in a mouse model of Alzheimer's disease. Our findings demonstrate a previously unknown neuroprotective mechanism in Alzheimer's disease, in which histone deacetylase 6 inhibition by valproic acid increases V-ATPase assembly and lysosomal acidification.
Collapse
Affiliation(s)
- Zhimin Long
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Chuanhua Ge
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Yueyang Zhao
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Yuanjie Liu
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Qinghua Zeng
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Qing Tang
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Zhifang Dong
- Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Guiqiong He
- Institute of Neuroscience, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Yao J, Li Y, Liu X, Liang W, Li Y, Wu L, Wang Z, Song W. FUBP3 mediates the amyloid-β-induced neuronal NLRP3 expression. Neural Regen Res 2025; 20:2068-2083. [PMID: 39254567 PMCID: PMC11691456 DOI: 10.4103/nrr.nrr-d-23-01799] [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: 11/01/2023] [Revised: 01/29/2024] [Accepted: 03/13/2024] [Indexed: 09/11/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00028/figure1/v/2024-09-09T124005Z/r/image-tiff Alzheimer's disease is characterized by deposition of amyloid-β, which forms extracellular neuritic plaques, and accumulation of hyperphosphorylated tau, which aggregates to form intraneuronal neurofibrillary tangles, in the brain. The NLRP3 inflammasome may play a role in the transition from amyloid-β deposition to tau phosphorylation and aggregation. Because NLRP3 is primarily found in brain microglia, and tau is predominantly located in neurons, it has been suggested that NLRP3 expressed by microglia indirectly triggers tau phosphorylation by upregulating the expression of pro-inflammatory cytokines. Here, we found that neurons also express NLRP3 in vitro and in vivo, and that neuronal NLRP3 regulates tau phosphorylation. Using biochemical methods, we mapped the minimal NLRP3 promoter and identified FUBP3 as a transcription factor regulating NLRP3 expression in neurons. In primary neurons and the neuroblastoma cell line Neuro2A, FUBP3 is required for endogenous NLRP3 expression and tau phosphorylation only when amyloid-β is present. In the brains of aged wild-type mice and a mouse model of Alzheimer's disease, FUBP3 expression was markedly increased in cortical neurons. Transcriptome analysis suggested that FUBP3 plays a role in neuron-mediated immune responses. We also found that FUBP3 trimmed the 5' end of DNA fragments that it bound, implying that FUBP3 functions in stress-induced responses. These findings suggest that neuronal NLRP3 may be more directly involved in the amyloid-β-to-phospho-tau transition than microglial NLRP3, and that amyloid-β fundamentally alters the regulatory mechanism of NLRP3 expression in neurons. Given that FUBP3 was only expressed at low levels in young wild-type mice and was strongly upregulated in the brains of aged mice and Alzheimer's disease mice, FUBP3 could be a safe therapeutic target for preventing Alzheimer's disease progression.
Collapse
Affiliation(s)
- Jing Yao
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuan Li
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xi Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Wenping Liang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yu Li
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liyong Wu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Weihong Song
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang Province, China
| |
Collapse
|
5
|
Qi G, Tang H, Hu J, Kang S, Qin S. Potential role of tanycyte-derived neurogenesis in Alzheimer's disease. Neural Regen Res 2025; 20:1599-1612. [PMID: 38934388 PMCID: PMC11688558 DOI: 10.4103/nrr.nrr-d-23-01865] [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: 01/17/2024] [Revised: 02/19/2024] [Accepted: 04/17/2024] [Indexed: 06/28/2024] Open
Abstract
Tanycytes, specialized ependymal cells located in the hypothalamus, play a crucial role in the generation of new neurons that contribute to the neural circuits responsible for regulating the systemic energy balance. The precise coordination of the gene networks controlling neurogenesis in naive and mature tanycytes is essential for maintaining homeostasis in adulthood. However, our understanding of the molecular mechanisms and signaling pathways that govern the proliferation and differentiation of tanycytes into neurons remains limited. This article aims to review the recent advancements in research into the mechanisms and functions of tanycyte-derived neurogenesis. Studies employing lineage-tracing techniques have revealed that the neurogenesis specifically originating from tanycytes in the hypothalamus has a compensatory role in neuronal loss and helps maintain energy homeostasis during metabolic diseases. Intriguingly, metabolic disorders are considered early biomarkers of Alzheimer's disease. Furthermore, the neurogenic potential of tanycytes and the state of newborn neurons derived from tanycytes heavily depend on the maintenance of mild microenvironments, which may be disrupted in Alzheimer's disease due to the impaired blood-brain barrier function. However, the specific alterations and regulatory mechanisms governing tanycyte-derived neurogenesis in Alzheimer's disease remain unclear. Accumulating evidence suggests that tanycyte-derived neurogenesis might be impaired in Alzheimer's disease, exacerbating neurodegeneration. Confirming this hypothesis, however, poses a challenge because of the lack of long-term tracing and nucleus-specific analyses of newborn neurons in the hypothalamus of patients with Alzheimer's disease. Further research into the molecular mechanisms underlying tanycyte-derived neurogenesis holds promise for identifying small molecules capable of restoring tanycyte proliferation in neurodegenerative diseases. This line of investigation could provide valuable insights into potential therapeutic strategies for Alzheimer's disease and related conditions.
Collapse
Affiliation(s)
- Guibo Qi
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Han Tang
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianian Hu
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Siying Kang
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Song Qin
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Huang J, Wu F, Cao W, Chen Y, Yao Q, Cen P, Wang J, Hong L, Zhang X, Zhou R, Jin C, Tian M, Zhang H, Zhong Y. Ultrasmall iron-gallic acid coordination polymer nanoparticles for scavenging ROS and suppressing inflammation in tauopathy-induced Alzheimer's disease. Biomaterials 2025; 317:123042. [PMID: 39805185 DOI: 10.1016/j.biomaterials.2024.123042] [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: 07/08/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder globally, with no effective treatment available yet. A crucial pathological hallmark of AD is the accumulation of hyperphosphorylated tau protein, which is deteriorated by reactive oxygen species (ROS) and neuroinflammation in AD progression. Thus, alleviation of ROS and inflammation has become a potential therapeutic strategy in many studies. Herein, we reported ultrasmall coordination polymer nanoparticles formed by ferric ions and gallic acid (Fe-GA CPNs), which owned antioxidant and anti-inflammation properties for AD therapeutics. The facilely prepared Fe-GA CPNs exhibited remarkable superoxide dismutase-like, peroxidase-like enzyme activity, and ROS eliminating ability with great water solubility, compared with gallic acid. We demonstrated that Fe-GA CPNs effectively relieved oxidative stress, ameliorated inflammation by modulating microglial polarization towards anti-inflammation phenotype, and reduced hyperphosphorylated tau protein levels. Furthermore, Fe-GA CPNs treatment significantly improved cognitive function in tauopathy-induced AD rats, and achieved a neuroprotective effect against AD pathology. This study highlights the potential of coordination polymer nanoparticles as promising therapeutic candidates for AD and other tau-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiani Huang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Fei Wu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Wenzhao Cao
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Yuhan Chen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Qiong Yao
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China
| | - Peili Cen
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Lu Hong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Xiaohui Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Chentao Jin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China; Human Phenome Institute, Fudan University, Shanghai, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China; Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China; College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China.
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China; Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China; Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| |
Collapse
|
7
|
Jain S, Murmu A, Chauhan A. Advancing Alzheimer's disease therapy through engineered exosomal Macromolecules. Brain Res 2025; 1855:149590. [PMID: 40120708 DOI: 10.1016/j.brainres.2025.149590] [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: 01/18/2025] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Exosomes are a subject of continuous investigation due to their function as extracellular vesicles (EVs) that significantly contribute to the pathophysiology of certain neurodegenerative disorders (NDD), including Alzheimer's disease (AD). Exosomes have shown the potential to carry both therapeutic and pathogenic materials; hence, researchers have used exosomes for medication delivery applications. Exosomes have reduced immunogenicity when used as natural drug delivery vehicles. This guarantees the efficient delivery of the medication without causing significant side reactions. Exosomes have lately enabled the potential for drug delivery in AD, along with promising future therapeutic uses for the detection of neurodegenerative disorders. Furthermore, exosomes have been examined for their prospective use in illness diagnosis and prediction before the manifestation of symptoms. This review will document prior studies and will concentrate on the rationale behind the substantial potential of exosomes in the treatment of AD and their prospective use as a diagnostic and predictive tool for this condition.
Collapse
Affiliation(s)
- Smita Jain
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Kishangarh, Rajasthan, India.
| | - Ankita Murmu
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Kishangarh, Rajasthan, India
| | - Aparna Chauhan
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Kishangarh, Rajasthan, India
| |
Collapse
|
8
|
Khandayataray P, Murthy MK. Exploring the nexus: Sleep disorders, circadian dysregulation, and Alzheimer's disease. Neuroscience 2025; 574:21-41. [PMID: 40189132 DOI: 10.1016/j.neuroscience.2025.03.066] [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: 01/02/2025] [Revised: 03/10/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
We reviewed the connections among Alzheimer's disease (AD), sleep deprivation, and circadian rhythm disorders. Evidence is mounting that disrupted sleep and abnormal circadian rhythms are not merely symptoms of AD, but are also involved in accelerating the disease. Amyloid-beta (Aβ) accumulates, a feature of AD, and worsens with sleep deprivation because glymphatic withdrawal is required to clear toxic proteins from the brain. In addition, disturbances in circadian rhythm can contribute to the induction of neuroinflammation and oxidative stress, thereby accelerating neurodegenerative processes. While these interactions are bidirectional, Alzheimer's pathology further disrupts sleep and circadian function in a vicious cycle that worsens cognitive decline, which is emphasized in the review. The evidence that targeting sleep and circadian mechanisms may serve as therapeutic strategies for AD was strengthened by this study through the analysis of the molecular and physiological pathways. Further work on this nexus could help unravel the neurobiological mechanisms common to the onset of Alzheimer's and disrupted sleep and circadian regulation, which could result in earlier intervention to slow or prevent the onset of the disease.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab 140401, India.
| |
Collapse
|
9
|
D'Alessandro MCB, Kanaan S, Geller M, Praticò D, Daher JPL. Mitochondrial dysfunction in Alzheimer's disease. Ageing Res Rev 2025; 107:102713. [PMID: 40023293 DOI: 10.1016/j.arr.2025.102713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by progressive cognitive decline and distinct neuropathological features. The absence of a definitive cure presents a significant challenge in neurology and neuroscience. Early clinical manifestations, such as memory retrieval deficits and apathy, underscore the need for a deeper understanding of the disease's underlying mechanisms. While amyloid-β plaques and tau neurofibrillary tangles have dominated research efforts, accumulating evidence highlights mitochondrial dysfunction as a central factor in AD pathogenesis. Mitochondria, essential cellular organelles responsible for energy production necessary for neuronal function become impaired in AD, triggering several cellular consequences. Factors such as oxidative stress, disturbances in energy metabolism, failures in the mitochondrial quality control system, and dysregulation of calcium release are associated with mitochondrial dysfunction. These abnormalities are closely linked to the neurodegenerative processes driving AD development and progression. This review explores the intricate relationship between mitochondrial dysfunction and AD pathogenesis, emphasizing its role in disease onset and progression, while also considering its potential as a biomarker and a therapeutic target.
Collapse
Affiliation(s)
- Maria Clara Bila D'Alessandro
- Universidade Federal Fluminense, Faculty of Medicine, Desembargador Athayde Parreiras road 100, Niterói, Rio de Janeiro, Brazil.
| | - Salim Kanaan
- Universidade Federal Fluminense, Faculty of Medicine, Department of Pathology, Marquês do Paraná road, 303, 2nd floor, Niterói, Rio de Janeiro, Brazil.
| | - Mauro Geller
- Unifeso, Department of Immunology and Microbiology, Alberto Torres avenue 111, Teresópolis, Rio de Janeiro, Brazil
| | - Domenico Praticò
- Department of Neurosciences, Lewis Katz School of Medicine. Temple University, 3500 North Broad Street, Philadelphia, PA, United States.
| | - João Paulo Lima Daher
- Universidade Federal Fluminense, Faculty of Medicine, Department of Pathology, Marquês do Paraná road, 303, 2nd floor, Niterói, Rio de Janeiro, Brazil.
| |
Collapse
|
10
|
Mukherjee AG, Mishra S, Gopalakrishnan AV, Kannampuzha S, Murali R, Wanjari UR, B S, Vellingiri B, Madhyastha H, Kanagavel D, Vijayan M. Unraveling the mystery of citrate transporters in Alzheimer's disease: An updated review. Ageing Res Rev 2025; 107:102726. [PMID: 40073978 DOI: 10.1016/j.arr.2025.102726] [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: 06/25/2024] [Revised: 12/26/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
A key molecule in cellular metabolism, citrate is essential for lipid biosynthesis, energy production, and epigenetic control. The etiology of Alzheimer's disease (AD), a progressive neurodegenerative illness marked by memory loss and cognitive decline, may be linked to dysregulated citrate transport, according to recent research. Citrate transporters, which help citrate flow both inside and outside of cells, are becoming more and more recognized as possible participants in the molecular processes underlying AD. Citrate synthase (CS), a key enzyme in the tricarboxylic acid (TCA) cycle, supports mitochondrial function and neurotransmitter synthesis, particularly acetylcholine (ACh), essential for cognition. Changes in CS activity affect citrate availability, influencing energy metabolism and neurotransmitter production. Choline, a precursor for ACh, is crucial for neuronal function. Lipid metabolism, oxidative stress reactions, and mitochondrial function can all be affected by aberrant citrate transport, and these changes are linked to dementia. Furthermore, the two main pathogenic characteristics of AD, tau hyperphosphorylation and amyloid-beta (Aβ) aggregation, may be impacted by disturbances in citrate homeostasis. The goal of this review is to clarify the complex function of citrate transporters in AD and provide insight into how they contribute to the development and course of the illness. We aim to provide an in-depth idea of which particular transporters are dysregulated in AD and clarify the functional implications of these dysregulated transporters in brain cells. To reduce neurodegenerative processes and restore metabolic equilibrium, we have also discussed the therapeutic potential of regulating citrate transport. Gaining insight into the relationship between citrate transporters and the pathogenesis of AD may help identify new indicators for early detection and creative targets for treatment. This study offers hope for more potent ways to fight this debilitating illness and is a crucial step in understanding the metabolic foundations of AD.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Shatakshi Mishra
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Stany B
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab 151401, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan
| | - Deepankumar Kanagavel
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
11
|
Tasnády KR, Jehoul R, de Ravé MG, Gijbels MJ, Brône B, Dewachter I, Melotte V, Boesmans W. Gastrointestinal Dysfunction and Low-Grade Inflammation Associate With Enteric Neuronal Amyloid-β in a Model for Amyloid Pathology. Neurogastroenterol Motil 2025; 37:e15016. [PMID: 40051115 PMCID: PMC11996054 DOI: 10.1111/nmo.15016] [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: 05/28/2024] [Revised: 12/18/2024] [Accepted: 01/20/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Patients suffering from Alzheimer's disease, a progressive neurodegenerative disorder involving cognitive decline and memory impairment, often present with gastrointestinal comorbidities. Accumulating data also indicate that alterations in the gut can modulate Alzheimer's disease pathology, highlighting the need to better understand the link between gastrointestinal abnormalities and neurodegeneration in the brain. METHODS To disentangle the pathophysiology of gastrointestinal dysfunction in Alzheimer's disease, we conducted a detailed pathological characterization of the gastrointestinal tract of 5xFAD mice by performing histological analyses, gene expression studies, immunofluorescence labeling and gut function assays. RESULTS We found that 5xFAD mice have elevated levels of intestinal amyloid precursor protein and accumulate amyloid-β in enteric neurons. Histopathology revealed that this is associated with mild intestinal inflammation and fibrosis and accompanied by increased expression of proinflammatory cytokines. While overall enteric nervous system composition and organization appeared unaffected, 5xFAD mice have faster gastrointestinal transit. CONCLUSION Our findings indicate that amyloid-β accumulation in enteric neurons is associated with low-grade intestinal inflammation and altered motility and suggest that peripheral pathology may cause gastrointestinal dysfunction in Alzheimer's disease patients.
Collapse
Affiliation(s)
- Kinga Réka Tasnády
- Biomedical Research Institute (BIOMED)Hasselt UniversityDiepenbeekBelgium
- Department of Pathology, GROW‐Research Institute for Oncology and ReproductionMaastricht University Medical CentreMaastrichtthe Netherlands
| | - Reindert Jehoul
- Biomedical Research Institute (BIOMED)Hasselt UniversityDiepenbeekBelgium
| | | | - Marion J. Gijbels
- Department of Pathology, NUTRIM Institute of Nutrition and Translational Research in MetabolismMaastricht University Medical CentreMaastrichtthe Netherlands
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Infection and Immunity, Amsterdam Cardiovascular SciencesAmsterdam University Medical CenterAmsterdamthe Netherlands
| | - Bert Brône
- Biomedical Research Institute (BIOMED)Hasselt UniversityDiepenbeekBelgium
| | - Ilse Dewachter
- Biomedical Research Institute (BIOMED)Hasselt UniversityDiepenbeekBelgium
| | - Veerle Melotte
- Department of Pathology, GROW‐Research Institute for Oncology and ReproductionMaastricht University Medical CentreMaastrichtthe Netherlands
- Department of Clinical GeneticsErasmus University Medical CenterRotterdamthe Netherlands
| | - Werend Boesmans
- Biomedical Research Institute (BIOMED)Hasselt UniversityDiepenbeekBelgium
- Department of Pathology, GROW‐Research Institute for Oncology and ReproductionMaastricht University Medical CentreMaastrichtthe Netherlands
| |
Collapse
|
12
|
Dastan M, Rajaei Z, Sharifi M, Salehi H. Crocin Improves Cognitive Impairment in LPS-treated Rats through Anti-Apoptotic, Anti-Inflammatory, and Antioxidant Activities. Mol Neurobiol 2025; 62:5804-5815. [PMID: 39630406 DOI: 10.1007/s12035-024-04638-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/18/2024] [Indexed: 03/29/2025]
Abstract
Brain inflammation and oxidative stress play critical roles in neuronal apoptosis and memory dysfunction in Alzheimer's disease. Crocin, a natural carotenoid in the stigma of saffron, possesses radical scavenging, anti-inflammatory, and anti-apoptotic properties. This study investigates the protective impact of crocin on neuronal apoptosis, oxidative stress, neuroinflammation, and memory deficits induced by lipopolysaccharide (LPS) in rats. Male Wistar rats received 100 mg/kg of crocin for 12 days, with LPS (1 mg/kg, ip) injected on days 8-12. Spatial learning and memory were evaluated in the Morris water maze two hours after LPS injection. Gene expression of nuclear factor kappa B (NF-κB), tumor necrosis factor-α (TNF-α), caspase 3, and lipid peroxidation was assessed in hippocampal homogenates at the end of the behavioral test. Histopathological changes in the hippocampus and cerebral cortex were evaluated using H&E staining. The results indicated that LPS administration caused spatial learning and memory dysfunction (P = 0.001, P < 0.01) accompanied by upregulation of Nfkb, Tnfα, and Casp3 mRNA expression (P < 0.0001), increased TNF-α (P < 0.01) and lipid peroxidation level (P < 0.01), decreased total thiol concentration (P < 0.05), tissue damage and neuronal loss in the hippocampus (P < 0.0001). Furthermore, crocin treatment at a dosage of 100 mg/kg attenuated learning and memory impairments (P = 0.001, P < 0.01), downregulated Nfkb, Tnfα, and Casp3 mRNA expression (P < 0.0001), decreased TNF-α level (P < 0.01) and lipid peroxidation (P < 0.05) and increased total thiol level (P < 0.05) in the hippocampus. Crocin also ameliorated LPS-induced pathological changes and neuronal loss in the hippocampus (P < 0.001) and cerebral cortex (P < 0.01). In conclusion, the neuroprotective effects of crocin against LPS-induced histopathological and behavioral changes could be attributed to its anti-apoptotic, anti-inflammatory, and radical-scavenging activities in the rat brain.
Collapse
Affiliation(s)
- Maryam Dastan
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ziba Rajaei
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
13
|
Zhang S, Liu H, Ouyang Z, Xu T, Yang Q, Zhu Y, Wan M, Xiao X, Yang X, Chen S, Yuan L, Bei Y, Wang J, Guo J, Chen H, Tang B, Luo S, Jiao B, Shen L. Accurate Diagnosis of Alzheimer's Disease Using Specific Breath Volatile Organic Compounds. ACS Sens 2025; 10:2699-2711. [PMID: 40107845 DOI: 10.1021/acssensors.4c03329] [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: 03/22/2025]
Abstract
Whether volatile organic compounds (VOCs) from exhaled breath can be used as a novel biomarker for Alzheimer's disease (AD) diagnosis is unclear. To determine the significantly distinctive VOCs for AD, a total of 970 participants were enrolled, including 60 individuals in data set 1 (AD, 30; controls, 30), 164 individuals in data set 2 (AD, 82; controls, 82), 637 individuals in data set 3 (AD, 31; controls, 606), and 109 individuals in data set 4 (frontotemporal dementia, 19; vascular dementia, 21; Parkinson's disease, 69). The participants in data sets 1, 2, and 4 were from Xiangya Hospital, Central South University. Participants in data set 3 were from a two-year follow-up cohort. VOCs in breath and plasma, neuropsychological scores, plasma p-tau181 levels, metabolites in plasma, and brain functional connectivity were detected. We found that six VOCs were significantly different between the two groups in data set 1 and were verified in data set 2 and data set 3. Ethanol (m/z = 46) and pyrrole (m/z = 67) presented AUC values of 0.907 and 0.895 in data sets 1 and 2 (clinical data sets) and 0.849 and 0.974 in data set 3 (community data set), respectively. The six VOCs were associated with cognitive decline as reflected by neuropsychological tests; five of them were correlated with plasma p-tau181, and these five plasma VOCs were consistently altered as breath VOCs. Correlation between metabolites and five VOCs in plasma was noted, and the five VOCs may originate from blood metabolites. Moreover, four breath VOCs were associated with altered brain connectivity. In conclusion, specific breath VOCs may be used as biomarkers for AD detection.
Collapse
Affiliation(s)
- Sizhe Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Haokun Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ziyu Ouyang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Tianyan Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qijie Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuan Zhu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Meidan Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xuewen Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xuan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shuliang Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Li Yuan
- Department of Neurology, Liuyang Jili Hospital, Changsha 410399, China
| | - Yuzhang Bei
- Department of Neurology, Liuyang Jili Hospital, Changsha 410399, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Haibin Chen
- Breax Laboratory, PCAB Research Center of Breath and Metabolism, Beijing 100000, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shilin Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Xiangya Hospital, Central South University, Changsha 410008, China
- Brain Research Center, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
14
|
Wang S, Qi C, Rajpurohit C, Ghosh B, Xiong W, Wang B, Qi Y, Hwang SH, Hammock BD, Li H, Gan L, Zheng H. Inhibition of soluble epoxide hydrolase confers neuroprotection and restores microglial homeostasis in a tauopathy mouse model. Mol Neurodegener 2025; 20:44. [PMID: 40264187 PMCID: PMC12016400 DOI: 10.1186/s13024-025-00844-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND The epoxyeicosatrienoic acids (EETs) are derivatives of the arachidonic acid metabolism with anti-inflammatory activities. However, their efficacy is limited due to the rapid hydrolysis by soluble epoxide hydrolase (sEH). Inhibition of sEH has been shown to stabilize the EETs and reduce neuroinflammation in Aβ mouse models of Alzheimer's disease (AD). However, the role of the sEH-EET signaling pathway in other CNS cell types and neurodegenerative conditions are less understood. METHODS Here we investigated the mechanisms and functional role of the sEH-EET axis in tauopathy by treating PS19 mice with a small molecule sEH inhibitor TPPU and by crossing the PS19 mice with Ephx2 (gene encoding sEH) knockout mice. This was followed by single-nucleus RNA-sequencing (snRNA-seq), biochemical and immunohistochemical analysis, and behavioral assessments. Additionally, we examined the effects of the sEH-EET pathway in primary microglia cultures and human induced pluripotent stem cell (iPSC)-derived neurons exhibiting seeding-induced Tau inclusions. RESULTS sEH inhibition improved cognitive function, rescued neuronal cell loss, and reduced Tau pathology and microglial reactivity. snRNA-seq revealed that TPPU treatment upregulated genes involved in actin cytoskeleton and excitatory synaptic pathways. Treatment of human iPSC-derived neurons with TPPU enhanced synaptic density without affecting Tau accumulation, suggesting a cell-autonomous neuroprotective effect of sEH blockade. Furthermore, sEH inhibition reversed disease-associated and interferon-responsive microglial states in PS19 mice, while EET supplementation promoted Tau phagocytosis and clearance in primary microglia cultures. CONCLUSION These findings demonstrate that sEH blockade or EET augmentation confers therapeutic benefit in neurodegenerative tauopathies by simultaneously targeting neuronal and microglial pathways.
Collapse
Affiliation(s)
- Shuo Wang
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Chuangye Qi
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Chetan Rajpurohit
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Baijayanti Ghosh
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Wen Xiong
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Baiping Wang
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yanyan Qi
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Hongjie Li
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
15
|
Dubbelman MA, Elias U, Palmer P, Dafni-Merom A, Gazit L, Udeogu OJ, Wang S, Papp KV, Amariglio RE, Arzy S, Marshall GA. Investigating the associations between tau and mental orientation among cognitively unimpaired individuals. J Alzheimers Dis 2025:13872877251334781. [PMID: 40267288 DOI: 10.1177/13872877251334781] [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: 04/25/2025]
Abstract
BackgroundImpairments in orientation in space, time, and person occur frequently in Alzheimer's disease (AD) dementia. Subtle changes in orientation may arise in preclinical and prodromal disease stages. Thus, assessing orientation may help identify those on a trajectory toward AD dementia.ObjectiveTo investigate how orientation, measured using a novel artificial intelligence-based paradigm, relates to AD biomarkers (amyloid and tau) in cognitively unimpaired older adults.MethodsUsing an automated chatbot, 53 cognitively unimpaired participants (74.0 ± 5.5 years; 60% female) provided details about memories and relationships, recognition of historical event dates, and geographical locations. These details were then used to assess orientation to space, time, and person. For each domain separately, orientation accuracy was calculated by dividing the number of correct responses by response time. All participants underwent Pittsburgh compound-B (amyloid) and flortaucipir (tau) positron emission tomography. We analyzed the relationship between performance on the three orientation domains and retrosplenial, precuneus, neocortical, and medial temporal tau, and global amyloid.ResultsHigher retrosplenial and precuneus tau burden were associated with worse temporal orientation (β = -0.32, 95% confidence interval [95%CI] = [-0.59, -0.05] and β = -0.29, 95%CI = [-0.57, -0.01], respectively). Spatial or social orientation were not associated with amyloid or tau.ConclusionsThese results suggest that impaired temporal orientation is related to AD pathological processes, even before the onset of overt cognitive impairment, and may infer a role for personalized assessment of orientation in early diagnosis of AD.
Collapse
Affiliation(s)
- Mark A Dubbelman
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Uri Elias
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| | - Phebe Palmer
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amnon Dafni-Merom
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| | - Lidor Gazit
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| | - Onyinye J Udeogu
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sharon Wang
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn V Papp
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca E Amariglio
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shahar Arzy
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| | - Gad A Marshall
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Yan X, Wang E, Zhao M, Ma G, Xu XX, Zhao JB, Li X, Zeng J, Ma X. Microbial infection instigates tau-related pathology in Alzheimer's disease via activating neuroimmune cGAS-STING pathway. Neuroscience 2025; 572:122-133. [PMID: 40064364 DOI: 10.1016/j.neuroscience.2025.03.019] [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: 06/23/2024] [Revised: 01/28/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Microbial infection, the strong trigger to directly induce inflammation in brain, is long considered a risk factor of Alzheimer's disease (AD), but how these infections contribute to neurodegeneration remains underexplored. To examine the effect of herpes simplex virus type 1 (HSV-1) infection on tauopathy in local hippocampus of P301S mice, we utilized a modified HSV-1 strain (mHSV-1) potentially relevant to AD, we found that its infection promotes tau-related pathology in part via activating neuroimmune cGAS-STING pathway in the tau mouse model. Specifically, Sting ablation causes the detectable improvement of neuronal dysfunction and loss in P301S mice, which is causally linked to lowered proinflammatory status in the brain. Administration of STING inhibitor H-151 alleviates neuroinflammation and tau-related pathology in P301S mice. These results jointly suggest that herpesviral infection, as the vital environmental risk factor, could induce tau-related pathology in AD pathogenesis partially via neuroinflammatory cGAS-STING pathway.
Collapse
Affiliation(s)
- Xiaoxu Yan
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Erlin Wang
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Meng Zhao
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Guanqin Ma
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Xiang-Xiong Xu
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Jie-Bin Zhao
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Xiaohong Li
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Jianxiong Zeng
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China; Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Yunnan Key Laboratory of Biodiversity Information, Kunming, Yunnan 650201, China.
| | - Xueling Ma
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
17
|
Chen J, Hadi F, Wen X, Zhao W, Xu M, Xue S, Lin P, Calandrelli R, Richard JLC, Song Z, Li J, Amani A, Liu Y, Chen X, Zhong S. Transcriptional regulation by PHGDH drives amyloid pathology in Alzheimer's disease. Cell 2025:S0092-8674(25)00397-6. [PMID: 40273909 DOI: 10.1016/j.cell.2025.03.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 02/06/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
Virtually all individuals aged 65 or older develop at least early pathology of Alzheimer's disease (AD), yet most lack disease-causing mutations in APP, PSEN, or MAPT, and many do not carry the APOE4 risk allele. This raises questions about AD development in the general population. Although transcriptional dysregulation has not traditionally been a hallmark of AD, recent studies reveal significant epigenomic changes in late-onset AD (LOAD) patients. We show that altered expression of the LOAD biomarker phosphoglycerate dehydrogenase (PHGDH) modulates AD pathology in mice and human brain organoids independent of its enzymatic activity. PHGDH has an uncharacterized role in transcriptional regulation, promoting the transcription of inhibitor of nuclear factor kappa-B kinase subunit alpha (IKKa) and high-mobility group box 1 (HMGB1) in astrocytes, which suppress autophagy and accelerate amyloid pathology. A blood-brain-barrier-permeable small-molecule inhibitor targeting PHGDH's transcriptional function reduces amyloid pathology and improves AD-related behavioral deficits. These findings highlight transcriptional regulation in LOAD and suggest therapeutic strategies beyond targeting familial mutations.
Collapse
Affiliation(s)
- Junchen Chen
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Fatemeh Hadi
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Xingzhao Wen
- Program in Bioinformatics and Systems Biology, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Wenxin Zhao
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Ming Xu
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Shuanghong Xue
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Pei Lin
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Riccardo Calandrelli
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | | | - Zhixuan Song
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Jessica Li
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Alborz Amani
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Yang Liu
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Xu Chen
- Department of Neurosciences, University of California, San Diego, La Jolla, San Diego, CA, USA; Neuroscience Graduate Program, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Sheng Zhong
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA, USA; Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA; Program in Bioinformatics and Systems Biology, University of California, San Diego, La Jolla, San Diego, CA, USA; Neuroscience Graduate Program, University of California, San Diego, La Jolla, San Diego, CA, USA.
| |
Collapse
|
18
|
Ashmawy RE, Okesanya OJ, Ukoaka BM, Daniel FM, Ezedigwe SG, Agboola AO, Ahmed MM, Ogaya JB, Amisu BO, Adigun OA, Oluwakemi OG, Hamza AM, Mourid MR, Kouwenhoven M, Lucero-Prisno DE. Exploring the efficacy and safety of lecanemab in the management of early Alzheimer's disease: A systematic review of clinical evidence. J Alzheimers Dis 2025:13872877251331640. [PMID: 40232258 DOI: 10.1177/13872877251331640] [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: 04/16/2025]
Abstract
BackgroundAlzheimer's disease (AD) is a growing neurodegenerative disorder causing cognitive decline, memory loss, and functional impairment. Lecanemab has shown safety and efficacy in clinical trials.ObjectiveThis review aims to understand the clinical evidence of lecanemab's effectiveness and safety in managing early AD.MethodsA systematic search was conducted using the Scopus database and ClinicalTrials.gov. Studies from 2014 to 2024 on lecanemab's safety, efficacy, and clinical outcomes for AD were included. Data extraction involved two independent reviewers, with synthesis using qualitative methodology.ResultsFindings from 13 studies and 13 ongoing clinical trials were reported, showing that lecanemab substantially reduces amyloid plaque load in the brains of AD patients. The therapeutic regimens vary across reported studies and trials, ranging from 2.5 mg/kg biweekly, 5 mg/kg monthly, 5 mg/kg biweekly, 10 mg/kg monthly, and 10 mg/kg intravenously biweekly. The Clarity AD phase 3 trial, the AHEAD study, and the DIAN-TU-001 trials have reported positive study outcomes with robust efficacy and safety outcomes with minimal side effects. Completed and ongoing trials report on the onset of amyloid-related imaging abnormalities (ARIA) and the continuation of care status following the onset of ARIA in these patients. The common infusion-related reactions were observed in 26.4% of the lecanemab group compared to 7% in the placebo group.ConclusionsThe management of AD has evolved over the years with the introduction of novel therapeutic agents like lecanemab. While its safety profile is generally favorable, careful monitoring is essential.
Collapse
Affiliation(s)
| | - Olalekan John Okesanya
- Department of Medical Laboratory Science, Neuropsychiatric Hospital, Abeokuta, Nigeria
- Faculty of Medicine, Department of Public Health and Maritime Transport, University of Thessaly, Volos, Greece
| | | | | | | | | | - Mohamed Mustaf Ahmed
- Faculty of Medicine and Health Sciences, SIMAD University, Mogadishu, Somalia
- Department of Research and Innovations, eHealth Somalia, Mogadishu, Somalia
| | - Jerico Bautista Ogaya
- Department of Medical Technology, Institute of Health Sciences and Nursing, Far Eastern University, Manila, Philippines
- Center for University Research, University of Makati, Makati City, Philippines
| | | | | | | | | | | | - Mbn Kouwenhoven
- Department of Physics, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Don Eliseo Lucero-Prisno
- Department of Global Health and Development, London School of Hygiene and Tropical Medicine, London, UK
- Research and Innovation Office, Southern Leyte State University, Sogod, Philippines
- The Office of Research, St. Paul University Iloilo, Iloilo City, Philippines
| |
Collapse
|
19
|
Wu M, Chen YF, Yao W, Zhou S, Xie Z, Tao Y, Zhong Y, Ma W. The anti-inflammatory drug Montelukast ameliorates cognitive deficits by rescuing the inflammatory levels in young AD animal models. Sci Rep 2025; 15:12720. [PMID: 40222965 PMCID: PMC11994820 DOI: 10.1038/s41598-025-91785-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 02/24/2025] [Indexed: 04/15/2025] Open
Abstract
Neuroinflammation precedes the clinical symptoms onset of Alzheimer's disease (AD) by decades. However, the anti-inflammatory drugs were not always effective at all stages of the disease. Here, using the fly and mouse AD models, we evaluated the effects of anti-inflammatory drugs on inflammatory-related factors and the proinflammatory cytokines at different ages of AD animals. We also performed behavioral tests to evaluate the cognitive aspects of AD. Combined with the bioinformatics analysis, we would like to exhibit a better understanding of AD. Based on the previous studies and reanalysis of published database, we found aged AD animals might better represent the inflammatory status of symptomatic AD. Our results showed that mRNA levels of antimicrobial peptides (AMPs) were highly expressed in 10-day-old AD flies, while no significant difference was observed in 40-day-old AD. In aged APP/PS1 mice (22.5 months), inflammatory-related factors NF-κB, IBA1, and the mRNA levels of proinflammatory cytokines Il-1β and Il-6 were not differentially expressed. In contrast, a significant increase was observed in 7.5-month-old APP/PS1 mice. Moreover, the anti-inflammatory drug Montelukast (MON) did not ameliorate the inflammatory and cognitive defects in 22.5-month-old aged mice but showed a rescue effect in 7.5-month-old young APP/PS1 mice. Altogether, our study demonstrates the different inflammatory status might lead to variations of anti-inflammatory drug efficacy, which helps to clarify the importance of considering the pathological stage of the disease when administering treatment.
Collapse
Affiliation(s)
- Mengnan Wu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yan-Fen Chen
- Beijing Joekai Biotechnology LLC, Beijing, 100094, China
| | - Wei Yao
- Beijing Joekai Biotechnology LLC, Beijing, 100094, China
| | - Siyan Zhou
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zuolei Xie
- Beijing Joekai Biotechnology LLC, Beijing, 100094, China
| | - Ye Tao
- Beijing Joekai Biotechnology LLC, Beijing, 100094, China
| | - Yi Zhong
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Weiwei Ma
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Beijing Joekai Biotechnology LLC, Beijing, 100094, China.
| |
Collapse
|
20
|
Ji HN, Zhou HQ, Qie JB, Lu WM, Gao HT, Wu DH. Dysregulated ac4C modification of mRNA in a mouse model of early-stage Alzheimer's disease. Cell Biosci 2025; 15:45. [PMID: 40223095 PMCID: PMC11995559 DOI: 10.1186/s13578-025-01389-8] [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: 11/26/2024] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND The identification and intervention of Alzheimer's Disease (AD) in its early-stage allows for the timely implementation of lifestyle modifications and therapeutic strategies. Although dysregulation of protein expression has been reported in the brain from AD patients and AD animal models, the underlying mechanisms remain poorly understood. N4-acetylcytidine (ac4C), the only known form of RNA acetylation in eukaryotes, has recently been shown to regulate mRNA stability and translation efficiency. However, the dysregulation of ac4C associated with abnormal protein expression levels in the brain of early-stage mouse models of AD remains to be elucidated. METHODS This study investigated ac4C modifications, mRNA and protein expression in the hippocampus of 3 and 6-month-old 5×FAD mice, a mouse model of AD, and wild-type (WT) littermates. The multi-omics analysis was performed: acetylated RNA immunoprecipitation followed by next-generation sequencing (acRIP-seq) to identify ac4C mRNAs, deep RNA sequencing (RNA-seq) to quantify mRNA abundance, and label-free quantitative proteomics to assess protein expression levels. In addition, we used acRIP-qPCR, regular qPCR and western blots to verify the ac4C, mRNA and protein levels of some key genes that were identified by the high-throughput assays. RESULTS Proteomic analysis revealed significant change of protein expression in the hippocampus of 3-months-old 5×FAD mice, compared with WT littermates. In contrast, RNA-seq analysis indicated that there were no substantial alterations in mRNA expression levels in the hippocampus of 3-months-old 5×FAD mice, compared to WT littermates. Strikingly, acRIP-seq revealed notable variations in ac4C modification on mRNAs, particularly those associated with synaptic structure and function, in the hippocampus of 3-months-old 5×FAD mice, compared with WT littermates. The ac4C modifications were found to be correlated with protein expression changes. Genes that are essential for synaptic function and cognition, including GRIN1, MAP2, and DNAJC6, exhibited reduced ac4C and protein levels in 3-months-old 5×FAD mice, without any corresponding changes in the mRNA levels, compared with WT littermates. Moreover, only a small part of dysregulated ac4C mRNAs identified in the 3-month-old 5×FAD mice were found in the 6-month-old 5×FAD mice. CONCLUSIONS Altogether these results identified abnormal ac4C modification of mRNAs that may contribute to the dysregulation of protein synthesis in the hippocampus from an early-stage mouse model of AD.
Collapse
Affiliation(s)
- Hao-Nan Ji
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Hai-Qian Zhou
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Jing-Bo Qie
- Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Wen-Mei Lu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Hai-Tao Gao
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Dan-Hong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China.
- Center of Community-Based Health Research, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
21
|
Wei C, Zhang G, Fu X, Zhao M, Zhai W, Shen Y, Sun L. Correlation of peripheral olfactomedin 1 with Alzheimer's disease and cognitive functions. Transl Psychiatry 2025; 15:146. [PMID: 40221403 PMCID: PMC11993663 DOI: 10.1038/s41398-025-03373-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/05/2024] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
Olfactomedin 1 (OLFM1) is thought to be involved in neuronal development, synaptic structure and function. However, the expression level of peripheral OLFM1 in Alzheimer's disease (AD) and its role in AD are unclear. The present study was conducted to assess the relationship of serum OLFM1 with AD and cognitive function. This study comprised 120 patients with AD and 118 healthy controls (HC). Serum OLFM1 levels, cognitive functions, and brain region volumes were evaluated in all participants. The results demonstrated a significant reduction in serum OLFM1 levels in AD patients (749.8 ± 42.3 pg/mL) compared to HC (804.4 ± 45.7 pg/mL). Among participants carrying the APOE ε4 allele, a significant positive correlation was observed between OLFM1 levels and cognitive assessments, including Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), and Memory and Executive Screening (MES). Furthermore, reduced OLFM1 levels were significantly associated with hippocampus (β = 0.005, 95% CI = 0.001-0.011, p = 0.042) and angular gyrus (β = 0.012, 95% CI = 0.001-0.022, p = 0.025) atrophy. The integration of serum OLFM1 with basic clinical characteristics exhibited robust discriminatory power in differentiating AD patients from HC, evidenced by an area under the curve of 0.881 (95% CI = 0.834-0.926). In summary, serum OLFM1 is a potential peripheral biomarker for AD, that correlates with cognitive function and specific brain volumes. In addition, APOE ε4 may modulate the influence of OLFM1 on cognitive function.
Collapse
Affiliation(s)
- Chunxiao Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiaoshu Fu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Meng Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Weijie Zhai
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yanxin Shen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China.
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
22
|
Cheng Y, Zhao F, Wang J, Luo H, Mao R, Yu Y, Wang Y, Tan J, Hao X, Wang Y. Preparation, activity, and mechanistic insights of processed Polygala tenuifolia glycoprotein in ameliorating Alzheimer's disease. Int J Biol Macromol 2025; 309:143069. [PMID: 40220839 DOI: 10.1016/j.ijbiomac.2025.143069] [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: 03/25/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
This study examined the efficacy and mechanism of Polygala tenuifolia glycoprotein (ZPG) in alleviating Alzheimer's disease (AD) for clinical application. ZPG's effects were tested in scopolamine hydrobromide-induced AD mice using behavioral, histological, and biomarker investigations. Additionally, 16S rDNA sequencing and lipidomics revealed ZPG's impact on gut microbiota and lipid metabolism, supported by pathway enrichment and correlation analyses. JNK pathway modulation was studied in vitro with purified and characterized ZPG-2. Results showed ZPG significantly improved cognitive deficits, reduced hippocampal pathology, and normalized APP, p-JNK, bax, and bcl-2 levels in AD mice. It also modulated gut microbiota and lipid metabolism, particularly glycerophospholipid pathways. ZPG-2 exhibited neuroprotective effects in Aβ25-35-induced PC12 cells by reducing apoptosis, inhibiting LDH release, and regulating oxidative stress and JNK activity. Structural analysis identified ZPG-2 as a 26 kDa glycoprotein with an O-linked glycopeptide bond and random coil conformation. Correlation analysis showed significant gut microbiota-AD biomarker relationships. These findings suggest ZPG may alleviate AD by reducing oxidative stress, inhibiting apoptosis, modulating gut microbiota, enhancing lipid metabolism, and suppressing the JNK signaling pathway. ZPG may be medicinal, however, more research is needed to validate its efficacy and mechanisms. This study lays the foundation for ZPG as an AD therapy for the future.
Collapse
Affiliation(s)
- Yangang Cheng
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Fuxia Zhao
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Jing Wang
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Hong Luo
- The University of Adelaide, Adelaide, SA 5005, Australia
| | - Rui Mao
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Yuetong Yu
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Yan Wang
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China
| | - Jinyan Tan
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China.
| | - Xuliang Hao
- Shanxi Academy of Traditional Chinese Medicine, Shanxi, Taiyuan 030024, China.
| | - Yingli Wang
- Shanxi University of Chinese Medicine, Shanxi, Jinzhong 030619, China.
| |
Collapse
|
23
|
Gong K, Zhou S, Xiao L, Xu M, Zhou Y, Lu K, Yu X, Zhu J, Liu C, Zhu Q. Danggui Shaoyao San ameliorates Alzheimer's disease by regulating lipid metabolism and inhibiting neuronal ferroptosis through the AMPK/Sp1/ACSL4 signaling pathway. Front Pharmacol 2025; 16:1588375. [PMID: 40271063 PMCID: PMC12014676 DOI: 10.3389/fphar.2025.1588375] [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: 03/05/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline; recent studies suggest that neuronal ferroptosis plays a key role in its pathogenesis. Danggui Shaoyao San (DSS), a traditional Chinese medicine formula, has shown demonstrated neuroprotective effects, but its precise mechanisms in AD treatment remain unclear. This study aims to investigate the mechanism of DSS in treating AD by inhibiting neuronal ferroptosis, explore whether DSS alleviates AD by suppressing neuronal ferroptosis via the AMPK/Sp1/ACSL4 pathway. Methods Chemical composition of DSS was identified by LC-MS/MS, followed by network pharmacology to predict targets and pathways. Molecular docking assessed binding affinities between DSS compounds and key proteins (AMPK, Sp1, ACSL4). In vivo experiments on APP/PS1 mice evaluated DSS effects on cognitive function, oxidative stress markers, lipid peroxidation, and ferroptosis-related proteins. Results Network pharmacology analysis suggested that DSS regulates lipid metabolism and inhibits neuronal ferroptosis via the AMPK pathway. Molecular docking revealed strong binding affinities between DSS compounds and AMPK downstream proteins, Sp1 and ACSL4. In vivo experiments showed that DSS improved cognitive function, enhanced antioxidant capacity, reduced lipid peroxide accumulation, and decreased Fe2+ content in brain tissue. Furthermore, DSS increased the expression of FTH, p-AMPK, and GPX4 while decreasing Sp1 and ACSL4 levels, thereby inhibiting ferroptosis. Conclusion DSS alleviates AD symptoms by suppressing neuronal ferroptosis via the AMPK/Sp1/ACSL4 axis, representing a novel lipid metabolism-targeted therapeutic strategy.
Collapse
Affiliation(s)
- Kai Gong
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuang Zhou
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Li Xiao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengzhen Xu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuhe Zhou
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kaihui Lu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xin Yu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiang Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingjun Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Jinan, China
| |
Collapse
|
24
|
Vosála O, Šmídová B, Novák J, Svoboda J, Petrásek T, Vojtěchová I, Macháček T. No evidence of Alzheimer's disease pathology in mice infected with Toxocara canis. Parasite 2025; 32:24. [PMID: 40214165 PMCID: PMC11987500 DOI: 10.1051/parasite/2025019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
The potential link between the infections and the development of Alzheimer's disease (AD) has led to speculations about the role of various pathogens in triggering amyloid-β (Aβ) overproduction, possibly leading to AD onset. The globally distributed dog roundworm Toxocara canis was suggested to be a suitable candidate due to neurotropism of the larvae and infection chronicity. This study investigated whether chronic T. canis infection induces AD-like pathology in mice and whether Aβ is toxic to T. canis. BALB/c and APP/PS1 transgenic mice, which overproduce Aβ, were infected with T. canis L3 larvae and monitored for larval burden, Aβ accumulation, and behavioral changes. In vitro tests of recombinant Aβ toxicity against the larvae were also performed. Despite the presence of T. canis larvae in the central nervous system 8 and 16 weeks post-infection, no significant increase in Aβ concentration or AD-related behavioral alterations were observed. Aβ was detected on the surface and within the intestines of T. canis larvae, but in vitro exposure to recombinant Aβ did not affect larval viability or morphology. Our findings suggest that T. canis infection does not trigger AD-like pathology in mice, and Aβ does not act as an antiparasitic agent. This challenges the emerging hypothesis that chronic neurotoxocarosis infections may contribute to AD development.
Collapse
Affiliation(s)
- Ondřej Vosála
- Department of Parasitology, Faculty of Science, Charles University Viničná 7 Prague 2 12844 Czechia
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 Hradec Králové 50005 Czechia
| | - Barbora Šmídová
- Department of Parasitology, Faculty of Science, Charles University Viničná 7 Prague 2 12844 Czechia
| | - Jan Novák
- Institute of Medical Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague Studničkova 7 Prague 2 12800 Czechia
| | - Jan Svoboda
- Laboratory of Neurophysiology of Memory, Institute of Physiology, Czech Academy of Sciences Vídeňská 1083 Prague 4 14200 Czechia
| | - Tomáš Petrásek
- Sleep and Chronobiology Research Centre, National Institute of Mental Health Topolová 748 Klecany 25067 Czechia
| | - Iveta Vojtěchová
- Sleep and Chronobiology Research Centre, National Institute of Mental Health Topolová 748 Klecany 25067 Czechia
| | - Tomáš Macháček
- Department of Parasitology, Faculty of Science, Charles University Viničná 7 Prague 2 12844 Czechia
| |
Collapse
|
25
|
Bi D, Bao H, Yang X, Wu Z, Yang X, Xu G, Liu X, Wan Z, Liu J, He J, Wen L, Jing Y, Zhu R, Long Z, Rong Y, Wang D, Wang X, Xiong W, Huang G, Gao F, Shen Y. BACE1-dependent cleavage of GABA A receptor contributes to neural hyperexcitability and disease progression in Alzheimer's disease. Neuron 2025; 113:1051-1064.e6. [PMID: 40015276 DOI: 10.1016/j.neuron.2025.01.030] [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: 06/01/2022] [Revised: 10/15/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025]
Abstract
Neural hyperexcitability has been clinically associated with amyloid-β (Aβ) pathology and cognitive impairment in Alzheimer's disease (AD). Here, we show that decreased GABAA receptor (GABAAR) currents are linked to hippocampal granule cell hyperexcitability in the AD mouse model APP23. Elevated levels of β-secretase (BACE1), the β-secretase responsible for generating Aβ peptides, lead to aberrant cleavage of GABAAR β1/2/3 subunits in the brains of APP23 mice and AD patients. Moreover, BACE1-dependent cleavage of the β subunits leads to a decrease in GABAAR-mediated inhibitory currents in BACE1 transgenic mice. Finally, we show that the neural hyperexcitability, Aβ load, and spatial memory deficit phenotypes of APP23 mice are significantly reduced upon the granule cell expression of a non-cleavable β3 subunit mutant. Collectively, our study establishes that BACE1-dependent cleavage of GABAAR β subunits promotes the pathological hyperexcitability known to drive neurodegeneration and cognitive impairment in the AD brain, suggesting that prevention of the cleavage could slow disease progression.
Collapse
Affiliation(s)
- Danlei Bi
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China.
| | - Hong Bao
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Xiaoli Yang
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Zujun Wu
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Xiaoxu Yang
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Guangwei Xu
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Xiaoming Liu
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Zhikun Wan
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Jiachen Liu
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Junju He
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Lang Wen
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Yuying Jing
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Ruijie Zhu
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Zhenyu Long
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Yating Rong
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Dongxu Wang
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Xiaoqun Wang
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Wei Xiong
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
| | - Guangming Huang
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Feng Gao
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China.
| | - Yong Shen
- Department of Neurology and 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, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
26
|
Jia M, Wang C, Mei J, Ahmad S, Nouman MF, Ai H. Identification and Characterization of the Structure and Size of Aβ42 Oligomers Targeting the Receptor FcγRIIb. ACS Chem Neurosci 2025; 16:1335-1345. [PMID: 40094208 DOI: 10.1021/acschemneuro.4c00862] [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: 03/19/2025] Open
Abstract
Kam and colleagues discovered that FcγRIIb can specifically bind to Aβ42 oligomers (AβOs). The N-terminal residues F4 and D7 of Aβ42, as well as the W115 residue in domain D2 of FcγRIIb, are involved in this binding. However, the specificity of the FcγRIIb receptor's binding sites for AβOs and their dependence on different AβO species, including dimers (D/DT), trimers (T/TT), tetramers (Te/TeT), and pentamers (P/PT) during both the primary (P1) and secondary nucleation phases (P2), remains unknown. To address this, we employed molecular dynamics (MD) simulations to investigate the interactions between the extracellular domains D1 and D2 (FDD) of FcγRIIb and AβOs of varying sizes in the two different phases. We discovered that three specific fragments (f1, f2, and f3) of domain D2 in FDD are the primary binding sites for AβO species. Furthermore, among AβOs of the same molecular weight, those from the P2 phase exhibit a stronger binding affinity for FDD than those from the P1 phase. The distinction is ascribed to the stronger dependence on the hydrophobic residues in the β1 and β2 regions for the binding of AβOs in P2 (including TT, TeT, and PT) than that (including D, Te, and P) in the P1 phase. In the P1 phase, these AβOs prefer to achieve binding to FDD through their N-terminal residues; however, by this, we identified that the species observed in Kam's experiment to bind FcγRIIb should probably be the tetrameric AβO (Te) in the P1 phase. Moreover, within both the P1 and P2 phases, we predicted that the trimeric AβO species in either the P1 or P2 phase is the strongest binding ligand for the FcγRIIb receptor. This study provides a comprehensive molecular perspective on the interaction between FcγRIIb and AβO in P2, which is of significant importance for the development of therapeutic strategies targeting Alzheimer's disease (AD) and autoimmune diseases.
Collapse
Affiliation(s)
- Mengke Jia
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
- Zibo City Engineering Research Center for New Pollution Monitoring and Governance, Shandong Vocational College of Light Industry, Zibo 255300, Shandong, P. R. China
| | - Chuanbo Wang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Jinfei Mei
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Sajjad Ahmad
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Muhammad Fahad Nouman
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Hongqi Ai
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| |
Collapse
|
27
|
Pang S, Li Z, Liu A, Luo ZH, Yin H, Fan S, Shi J, Liu N, Pan S, Yang YJ, Zhang GJ, Chen J. A Novel Oxo-Palmatine Derivative 2q as Potent Reversal Agents Against Alzheimer's Disease. Drug Dev Res 2025; 86:e70073. [PMID: 40079275 DOI: 10.1002/ddr.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/14/2025] [Accepted: 03/02/2025] [Indexed: 03/15/2025]
Abstract
Palmatine (PAL), as an active ingredient in traditional Chinese medicine, had been demonstrated efficacy in ameliorating the manifestations of AD. Our research group has previously designed and synthesized the novel oxo-PAL derivative 2q and found that it has exhibited notable neuroprotective properties. However, compound 2q therapeutic impact on AD remains uncertain. In the current investigation, our findings demonstrated that compound 2q displayed significant anti-AβOs activity in vitro by using xCELLigence analysis, and showed a high likelihood of crossing the blood-brain barrier. Furthermore, administration of compound 2q yielded a notable amelioration in Aβ accumulation and hyperphosphorylation of Tau in 3×Tg mice. Additionally, it was observed that compound 2q potentially enhanced the pathological characteristics of AD by targeting Potassium/Sodium Hyperpolarization-Activated Cyclic Nucleotide-Gated Channel 2 (HCN2). In conclusion, compound 2q emerged as a promising candidate for AD treatment, as it effectively restored AD-associated pathological impairments. Furthermore, it has been identified as a potential target of HCN2, thereby offering novel avenues for the development of treatments for AD.
Collapse
Affiliation(s)
- Shuo Pang
- The Laboratory of Neurological Disorders and Brain Cognition, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Zhuo Li
- Department of Neuropathology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ao Liu
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhuo-Hui Luo
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Heqing Yin
- The Laboratory of Neurological Disorders and Brain Cognition, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Songqiao Fan
- The Laboratory of Neurological Disorders and Brain Cognition, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Junjie Shi
- The Laboratory of Neurological Disorders and Brain Cognition, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Ning Liu
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuo Pan
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ya-Jun Yang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guo-Jun Zhang
- The Laboratory of Neurological Disorders and Brain Cognition, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Jun Chen
- The Laboratory of Neurological Disorders and Brain Cognition, Beijing Pediatric Research Institute, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Zhao Y, Jia M, Ding C, Bao B, Li H, Ma J, Dong W, Gao R, Chen X, Chen J, Dai X, Zou Y, Hu J, Shi L, Liu X, Liu Z. Time-restricted feeding mitigates Alzheimer's disease-associated cognitive impairments via a B. pseudolongum-propionic acid-FFAR3 axis. IMETA 2025; 4:e70006. [PMID: 40236783 PMCID: PMC11995186 DOI: 10.1002/imt2.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 04/02/2025]
Abstract
Time-restricted feeding (TRF) holds promise for alleviating cognitive decline in aging, albeit the precise mechanism via the gut-brain axis remains elusive. In a clinical trial, we observed, for the first time, that a 4-month TRF ameliorated cognitive impairments among Alzheimer's disease (AD) patients. Experiments in 5xFAD mice corroborated the gut microbiota-dependent effect of TRF on mitigating cognitive dysfunction, amyloid-beta deposition, and neuroinflammation. Multi-omics integration linked Bifidobacterium pseudolongum (B. pseudolongum) and propionic acid (PA) with key genes in AD pathogenesis. Oral supplementation of B. pseudolongum or PA mimicked TRF's protective effects. Positron emission tomography imaging confirmed PA's blood-brain barrier penetration, while knockdown of the free fatty acid receptor 3 (FFAR3) diminished TRF's cognitive benefits. Notably, we observed a positive correlation between fecal PA and improved cognitive function in an AD cohort, further indicating that TRF enhanced PA production. These findings highlight the microbiota-metabolites-brain axis as pivotal in TRF's cognitive benefits, proposing B. pseudolongum or PA as potential AD therapies.
Collapse
Affiliation(s)
- Yihang Zhao
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingChina
| | - Mengzhen Jia
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingChina
| | - Chen Ding
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingChina
| | - Bingkun Bao
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingChina
| | - Hangqi Li
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingChina
| | - Jiabin Ma
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingChina
| | - Weixuan Dong
- The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Rui Gao
- The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Xuhui Chen
- Peking University Shenzhen HospitalShenzhenChina
| | - Jiao Chen
- Peking University Shenzhen HospitalShenzhenChina
| | | | | | - Jun Hu
- Peking University Shenzhen HospitalShenzhenChina
| | - Lin Shi
- Shaanxi Normal UniversityXi'anChina
| | - Xuebo Liu
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingChina
| | - Zhigang Liu
- College of Food Science and EngineeringNorthwest A&F UniversityYanglingChina
- Northwest A&F University Shenzhen Research InstituteShenzhenChina
| |
Collapse
|
29
|
Kehs Z, Cross AC, Li YM. From defense to disease: IFITM3 in immunity and Alzheimer's disease. Neurotherapeutics 2025; 22:e00482. [PMID: 39516072 DOI: 10.1016/j.neurot.2024.e00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Innate immunity protein interferon induced transmembrane protein 3 (IFITM3) is a transmembrane protein that has a wide array of functions, including in viral infections, Alzheimer's Disease (AD), and cancer. As an interferon stimulated gene (ISG), IFITM3's expression is upregulated by type-I, II, and III interferons. Moreover, the antiviral activity of IFITM3 is modulated by post-translational modifications. IFITM3 functions in innate immunity to disrupt viral fusion and entry to the plasma membrane as well as prevent viral escape from endosomes. As a γ-secretase modulatory protein, IFITM3 distinctly modulates the processing of amyloid precursor protein (APP) to generate amyloid beta peptides (Aβ) and Notch1 cleavages. Increased IFITM3 expression, which can result from aging, cytokine activation, inflammation, and infection, can lead to an upregulation of γ-secretase for Aβ production that causes a risk of AD. Therefore, the prevention of IFITM3 upregulation has potential in the development of novel therapies for the treatment of AD.
Collapse
Affiliation(s)
- Zoe Kehs
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Abigail C Cross
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Programs of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA.
| |
Collapse
|
30
|
Hard SAAA, Shivakumar HN, Bafail DA, Moqbel Redhwan MA. Development of in vitro and in vivo evaluation of mucoadhesive in-situ gel for intranasal delivery of vinpocetine. J Drug Target 2025; 33:528-545. [PMID: 39601452 DOI: 10.1080/1061186x.2024.2433557] [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: 06/13/2024] [Revised: 09/21/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
ABSTRACT Alzheimer's disease (AD), which is marked by gradual neuronal decline and subsequent loss of cognitive functions and memory, poses significant treatment challenges. The present study involved the development, in vitro, and in vivo evaluation of a novel intranasal mucoadhesive in-situ gel of vinpocetine (VIN) with the aim to target the brain. An innovative gel formulation composed of poloxamer 407, HPMC E15 LV, and citric acid as a solubilizer was developed by 23 Factorial Design. The developed optimal formulation exhibited favorable rheological properties as it displayed ideal gelation time (31.6 ± 1.52 sec), optimum gelling temperature (32 ± 1.0 °C), enhanced mucoadhesive strength (6622 ± 2.64 dynes/cm2), prolonged adhesion (7.22 ± 0.57 hrs) compared with the baseline formulation (F18), and improved drug release in 12 hrs (39.59 ± 1.6%). In vivo, pharmacokinetics revealed a significant increase in Cmax (∼2-fold) and AUC0-t (∼2-fold) in the brain with the in-situ intranasal gel compared to the oral route. In the rat model of AD, in-situ intranasal gel demonstrated significantly greater efficacy (p < 0.001) than oral administration in alleviating AD symptoms as evidenced by behavioral and histological studies. Thus, VIN in-situ gel can be safe and noninvasive for nose-to-brain drug delivery.
Collapse
Affiliation(s)
- Sumaia Abdulbari Ahmed Ali Hard
- Department of Pharmaceutics, KLE College of Pharmacy, Bengaluru, Karnataka, India
- Basic Science Research Center (Off-Campus), KLE College of Pharmacy, Bengaluru, Karnataka, India
| | - H N Shivakumar
- Department of Pharmaceutics, KLE College of Pharmacy, Bengaluru, Karnataka, India
- Basic Science Research Center (Off-Campus), KLE College of Pharmacy, Bengaluru, Karnataka, India
| | - Duaa Abdullah Bafail
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Moqbel Ali Moqbel Redhwan
- Basic Science Research Center (Off-Campus), KLE College of Pharmacy, Bengaluru, Karnataka, India
- Department of Pharmacology, KLE College of Pharmacy, Bengaluru, Karnataka, India
| |
Collapse
|
31
|
Fawzy MN, Abd El-Haleim EA, Zaki HF, Salem HA, El-Sayed RM. Mitigating seizure-induced cognitive deficits in mice induced with pentylenetetrazol by roflumilast through targeting the NLRP3 inflammasome/BDNF/SIRT3 pathway and regulating ferroptosis. Life Sci 2025; 366-367:123488. [PMID: 39983820 DOI: 10.1016/j.lfs.2025.123488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
AIMS Comorbidities with epilepsy and antiseizure medications (ASMs) are currently the main challenges in treating epilepsy. The current study evaluates for the first time the neuroprotective effect of roflumilast (ROF) alone or combined with phenytoin (PHT) against pentylenetetrazol (PTZ)-induced kindling in mice. It focuses on the crosstalk between the NOD-like receptor protein 3 (NLRP3)/caspase 1/interleukin 1β (IL-1β) cascade and the brain-derived neurotrophic factor (BDNF)/sirtuin 3 (SIRT3) pathway as possible strategies to treat epilepsy. MAIN METHODS The kindled mouse model was induced via fifteen (35 mg/kg) intraperitoneal injections every other day. Roflumilast (0.4 mg/kg) and phenytoin (30 mg/kg) were orally administered daily from the start until the end of the experiment. Following the PTZ injection, the seizure severity score was assessed. The Morris water maze (MWM) test was performed to evaluate cognition. Histopathological examinations of hippocampi were conducted. KEY FINDINGS Roflumilast significantly improved neurobehavioral and histological assessments, whereas Racine scores declined. The improvement was confirmed through BDNF upregulation in contrast to NLRP3 and caspase-1 in the hippocampus, as revealed immunohistochemically. In addition, roflumilast induced a prominent elevation in gamma-aminobutyric acid (GABA), sirtuin 3 (SIRT3), and glutathione peroxidase (GPX4), whereas malondialdehyde (MDA), and arachidonic acid 15-lipoxygenase (ALOX15) expressions were downregulated. SIGNIFICANCE Our findings demonstrate that roflumilast conferred neuroprotective benefits against PTZ-induced kindling seizures, suggesting its potential as a novel adjuvant therapy for epilepsy-related disorders. This effect might be due to the modification of the NLRP3 inflammasome/BDNF pathway, ferroptosis, and a decrease in oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Mohamed N Fawzy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish 45511, Egypt.
| | - Enas A Abd El-Haleim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hesham A Salem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rehab M El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish 45511, Egypt
| |
Collapse
|
32
|
Wu J, Xu W, Su Y, Wang GH, Ma JJ. Targeting chaperone-mediated autophagy in neurodegenerative diseases: mechanisms and therapeutic potential. Acta Pharmacol Sin 2025; 46:816-828. [PMID: 39548290 PMCID: PMC11950187 DOI: 10.1038/s41401-024-01416-3] [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: 07/03/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
The pathological hallmarks of various neurodegenerative diseases including Parkinson's disease and Alzheimer's disease prominently feature the accumulation of misfolded proteins and neuroinflammation. Chaperone-mediated autophagy (CMA) has emerged as a distinct autophagic process that coordinates the lysosomal degradation of specific proteins bearing the pentapeptide motif Lys-Phe-Glu-Arg-Gln (KFERQ), a recognition target for the cytosolic chaperone HSC70. Beyond its role in protein quality control, recent research underscores the intimate interplay between CMA and immune regulation in neurodegeneration. In this review, we illuminate the molecular mechanisms and regulatory pathways governing CMA. We further discuss the potential roles of CMA in maintaining neuronal proteostasis and modulating neuroinflammation mediated by glial cells. Finally, we summarize the recent advancements in CMA modulators, emphasizing the significance of activating CMA for the therapeutic intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Jin Wu
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China.
| | - Wan Xu
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China
| | - Ying Su
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China
| | - Guang-Hui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Jing-Jing Ma
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
33
|
Sun M, Cai X, Lan Z, Liu M, Zhou M, Tang Y, Liu Y, Zhang X, Zhao X, Zhou Y, Zhang J, Meng Z. The lysosomal-associated membrane protein 2-macroautophagy pathway is involved in the regulatory effects of hippocampal aromatase on Aβ accumulation and AD-like behavior. Life Sci 2025; 366-367:123484. [PMID: 39983826 DOI: 10.1016/j.lfs.2025.123484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/19/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
AIMS Hippocampal aromatase (AROM) knockdown induces Aβ accumulation and Alzheimer's disease (AD)-like spatial learning and memory impairment, and early hippocampal AROM overexpression in APP/PS1 mice prevents Aβ deposition and memory loss later in life. The aim of this study was to elucidate the underlying mechanism and provide novel prevention and treatment targets for AD. MATERIALS AND METHODS AROM-inhibiting viral vectors were constructed and injected into the hippocampi of adult female mice, after which label-free LC-MS/MS proteomics and bioinformatics analysis were conducted. Additional viral vectors targeting LAMP2 or LC3 were constructed and used to treat HT22 cells. LAMP2 expression was verified, and macroautophagy levels, autophagosome formation and Aβ accumulation were examined. Additionally, ovariectomy combined with the hippocampal injection of LAMP2 inhibition/overexpression viral vectors was applied, and learning and memory abilities and Aβ accumulation were examined. KEY FINDINGS Proteomics revealed the enrichment of CMA and autophagy, and LAMP2 was the most significantly upregulated protein. Higher LAMP2 levels were correlated with lower macroautophagy and autophagosomes levels but were correlated with higher Aβ accumulation, and vice versa. Additionally, hippocampal LAMP2 mediated the effects of ovariectomy on spatial memory and Aβ accumulation. SIGNIFICANCE These results demonstrated the important role of the hippocampal LAMP2-macroautophagy pathway in mediating both hippocampal and ovarian estrogen regulation of Aβ accumulation and AD-like behavior, indicating that LAMP2 might be a novel target for both hippocampal and circulating estrogen deficiency-associated memory impairments, such as AD.
Collapse
Affiliation(s)
- Mingguang Sun
- Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing University of Chinese Medicine, Beijing 100853, China; Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Xiaoxia Cai
- Department of Neurobiology, Army Medical University, Chongqing 400038, China; School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Zhen Lan
- Department of Neurobiology, Army Medical University, Chongqing 400038, China; Department of General Surgery, General Hospital of Central Theater Command, Wuhan 430000, China
| | - Mengying Liu
- Department of Neurobiology, Army Medical University, Chongqing 400038, China; The 305 Hospital of PLA, Beijing 100017, China
| | - Maohu Zhou
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Yisha Tang
- College of Letters and Science, University of California, Berkeley, CA 94720, United States
| | - Yan Liu
- School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xuan Zhang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Xiao Zhao
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Yue Zhou
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Jiqiang Zhang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China.
| | - Zhaoyou Meng
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| |
Collapse
|
34
|
Ren Y, Pieper AA, Cheng F. Utilization of precision medicine digital twins for drug discovery in Alzheimer's disease. Neurotherapeutics 2025; 22:e00553. [PMID: 39965994 DOI: 10.1016/j.neurot.2025.e00553] [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: 10/16/2024] [Revised: 01/11/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Alzheimer's disease (AD) presents significant challenges in drug discovery and development due to its complex and poorly understood pathology and etiology. Digital twins (DTs) are recently developed virtual real-time representations of physical entities that enable rapid assessment of the bidirectional interaction between the virtual and physical domains. With recent advances in artificial intelligence (AI) and the growing accumulation of multi-omics and clinical data, application of DTs in healthcare is gaining traction. Digital twin technology, in the form of multiscale virtual models of patients or organ systems, can track health status in real time with continuous feedback, thereby driving model updates that enhance clinical decision-making. Here, we posit an additional role for DTs in drug discovery, with particular utility for complex diseases like AD. In this review, we discuss salient challenges in AD drug development, including complex disease pathology and comorbidities, difficulty in early diagnosis, and the current high failure rate of clinical trials. We also review DTs and discuss potential applications for predicting AD progression, discovering biomarkers, identifying new drug targets and opportunities for drug repurposing, facilitating clinical trials, and advancing precision medicine. Despite significant hurdles in this area, such as integration and standardization of dynamic medical data and issues of data security and privacy, DTs represent a promising approach for revolutionizing drug discovery in AD.
Collapse
Affiliation(s)
- Yunxiao Ren
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland 44106, OH, USA; Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA.
| |
Collapse
|
35
|
Kalamara TV, Dodos K, Georgakopoulou VE, Fotakopoulos G, Spandidos DA, Kapoukranidou D. Cognitive efficacy of omega‑3 fatty acids in Alzheimer's disease: A systematic review and meta‑analysis. Biomed Rep 2025; 22:62. [PMID: 39991006 PMCID: PMC11843191 DOI: 10.3892/br.2025.1940] [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: 08/22/2024] [Accepted: 02/03/2025] [Indexed: 02/25/2025] Open
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia, is a progressive neurodegenerative disorder characterized by a gradual decline in several domains of higher cortical function. Both preclinical and clinical research has suggested that the supplementation omega-3 fatty acids (FAs) may have potential benefits for individuals with AD. The present study aimed to identify all randomized controlled trials (RCTs) examining the association between omega-3 FA supplementation and cognitive function in patients with AD, using the Alzheimer's Disease Assessment Scale-Cognitive (ADAS-Cog) Subscale test as the primary outcome measure. A comprehensive search of the PubMed and Cochrane Library databases was conducted for all published RCTs up to December, 2023 that assessed cognition following omega-3 FA supplementation compared to placebo. A total of five studies met the eligibility criteria and were included in the qualitative synthesis, with four of these studies being incorporated into the meta-analysis. From these studies, data were collected from a total of 702 patients with AD, with 376 participants receiving omega-3 FA supplementation and 326 participants receiving a placebo. The primary outcome measure was the ADAS-Cog score. The meta-analysis revealed that omega-3 FA supplementation had a non-significant impact on the ADAS-Cog score compared to placebo, with a mean difference of 1.37 [95% confidence interval (CI) 0.00-2.73]. The heterogeneity among the included studies was moderate (I2=35%, P=0.17). The test for overall effect (z=1.96, P=0.05) indicated no statistical significance. Therefore, it was concluded that omega-3 FA supplementation does not significantly affect the cognitive function of adults with AD.
Collapse
Affiliation(s)
- Tsampika Vasileia Kalamara
- Laboratory of Physiology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Konstantinos Dodos
- Laboratory of Physiology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | | - George Fotakopoulos
- Department of Neurosurgery, General University Hospital of Larissa, 41221 Larissa, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dorothea Kapoukranidou
- Laboratory of Physiology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
36
|
Zhang L, Liu Y, Wang X, Wu H, Xie J, Liu Y. Treadmill exercise ameliorates hippocampal synaptic injury and recognition memory deficits by TREM2 in AD rat model. Brain Res Bull 2025; 223:111280. [PMID: 40015348 DOI: 10.1016/j.brainresbull.2025.111280] [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: 06/14/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVE The impairment of cognitive function has been associated with Alzheimer's disease (AD). Exercise exerts a positive modulatory effect on cognition by reducing synapse injury. However, limited in vivo evidence is available to validate the neuroprotective effect of TREM2 on synaptic function in this phenomenon. Here, we aim to explore whether physical exercise pretreatment alters Aβ-induced recognition memory impairment in structural synaptic plasticity within the hippocampus in AD rats. METHODS:: In study 1, fifty-two Sprague-Dawley (SD) rats were randomly divided into following four groups: control group (C group, n = 13), Alzheimer's disease group (AD group, n = 13), 4 weeks of physical exercise and Alzheimer's disease group (Exe+AD group, n = 13), 4 weeks of physical exercise and blank group (Exercise group, n = 13). Four weeks of treadmill exercise intervention was performed, and AD model were established by intra-cerebroventricular injection (ICV) injection of Aβ1-42 protein. After 3 weeks, we also conducted a novel object test to evaluate recognition memory in the behavior assessment. Golgi staining and transmission electron microscopy were used to evaluate the morphology and synaptic ultrastructure of neurons. Western blotting was used to measure the expression of hippocampal synaptic proteins. Extracellular neurotransmitters in the hippocampus were detected by microdialysis coupled with high-performance liquid chromatography. In study 2, 33 SD rats were randomly divided into three groups: 4 weeks of physical exercise and Alzheimer's disease group (Exe+AD group, n = 11), AAV-Control and physical exercise and Alzheimer's disease group (AAV-Control+Exe+AD group, n = 11), AAV-TREM2 and physical exercise and Alzheimer's disease group (AAV-TREM2 +Exe+AD group, n = 11). Stereotactic intracerebral injection in the bilateral hippocampus was performed to achieve microglial TREM2 down-expression by using adeno-associated virus (AAV) with CD68 promoter. After 4 weeks treadmill exercise and 3 weeks Aβ injection, all rats received behavior test and molecular experiment, which the same with experiment 2. RESULTS Novel recognition index in novel object recognition test significantly decreased, and western blot demonstrate that hippocampal TREM2 protein is significantly decreased (P < 0.001). But physical exercise reversed this phenomenon(P < 0.001). In addition, compared with Con group, the neuron from Exe+AD group exhibited a more complex branching pattern (P < 0.05). And impaired synaptic ultrastructure was observed in AD group. Hippocampal synaptic-related protein (SYX, SYP, GAP43, PSD95) and neurotransmitter (DA, Glu, GABA) was also significantly decreased (P < 0.01) in AD group. But the neuroprotection effect can be found in Exe+AD group, which are associated with the inhibition of synaptic injury by activate hippocampal TREM2 (P < 0.05). However, when blockade of hippocampal TREM2 reduced brain protective effect of exercise in AD rat model, including increased the damage of neuronal dendritic complexity, synaptic ultrastructure, and the decrease of hippocampal synapses-related protein, typical neurotransmitter. CONCLUSION Treadmill exercise facilitated recognition memory acquisition via TREM2-mediated structural synaptic plasticity of the hippocampus in an AD rat model.
Collapse
Affiliation(s)
- Linlin Zhang
- Department of physical education, Henan normal university, Xinxiang 453007, China; Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Yanzhong Liu
- School of physical education and health, Henan University of China Medicine, Zhengzhou, China
| | - Xin Wang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Hao Wu
- Comprehensive Key Laboratory of Sports Ability Evaluation and Research of the General Administration of Sport of China, Beijing Key Laboratory of Sports Function Assessment and Technical Analysis, Capital University of Physical Education and Sports, Beijing 100191, China
| | - Jiahui Xie
- Department of Physical Education and Research, Fuzhou University, Fuzhou 350108, China.
| | - Yiping Liu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou 350007, China.
| |
Collapse
|
37
|
Wang X, Feng S, Deng Q, Wu C, Duan R, Yang L. The role of estrogen in Alzheimer's disease pathogenesis and therapeutic potential in women. Mol Cell Biochem 2025; 480:1983-1998. [PMID: 39088186 DOI: 10.1007/s11010-024-05071-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
Estrogens are pivotal regulators of brain function throughout the lifespan, exerting profound effects from early embryonic development to aging. Extensive experimental evidence underscores the multifaceted protective roles of estrogens on neurons and neurotransmitter systems, particularly in the context of Alzheimer's disease (AD) pathogenesis. Studies have consistently revealed a greater risk of AD development in women compared to men, with postmenopausal women exhibiting heightened susceptibility. This connection between sex factors and long-term estrogen deprivation highlights the significance of estrogen signaling in AD progression. Estrogen's influence extends to key processes implicated in AD, including amyloid precursor protein (APP) processing and neuronal health maintenance mediated by brain-derived neurotrophic factor (BDNF). Reduced BDNF expression, often observed in AD, underscores estrogen's role in preserving neuronal integrity. Notably, hormone replacement therapy (HRT) has emerged as a sex-specific and time-dependent strategy for primary cardiovascular disease (CVD) prevention, offering an excellent risk profile against aging-related disorders like AD. Evidence suggests that HRT may mitigate AD onset and progression in postmenopausal women, further emphasizing the importance of estrogen signaling in AD pathophysiology. This review comprehensively examines the physiological and pathological changes associated with estrogen in AD, elucidating the therapeutic potential of estrogen-based interventions such as HRT. By synthesizing current knowledge, it aims to provide insights into the intricate interplay between estrogen signaling and AD pathogenesis, thereby informing future research directions and therapeutic strategies for this debilitating neurodegenerative disorder.
Collapse
Affiliation(s)
- Xinyi Wang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Shu Feng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
38
|
Yin X, Sun J, Wang X, Wu W, Chen Z, Zhang D, Xu Y, Chen Y, Qiu W, Qian X, Ni J, Ma C. Prevalence of cerebral amyloid angiopathy and its correlation with Alzheimer's disease and cognition in an autopsy-confirmed cohort from China. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70100. [PMID: 40201593 PMCID: PMC11973253 DOI: 10.1002/dad2.70100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND We aimed to investigate the prevalence of cerebral amyloid angiopathy (CAA) and its correlations with Alzheimer's disease (AD) and cognitive impairment in an autopsy-confirmed cohort donated to a human brain bank in Beijing, China. METHODS A total of 483 subjects were neuropathologically evaluated based on standardized protocols. Descriptive statistics and ordinal logistic regression models were used to estimate the correlation between CAA, AD, apolipoprotein E (APOE) genotyping, and cognitive function proximal to death. RESULTS Neuropathological assessment revealed that 53 of 483 subjects (11%) had CAA without AD, 78 of 483 (16%) had AD without CAA, 98 of 483 (20%) had both CAA and AD, and 254 of 483 (53%) had neither condition. A significant correlation was confirmed between CAA severity and AD. Subjects with both CAA and AD exhibited aggravated cognitive impairment. DISCUSSION Our results indicate a substantial prevalence of CAA that is frequently comorbid with AD and may exacerbate cognitive decline in the elderly population in China. Highlights First reporting of cerebral amyloid angiopathy (CAA) based on an autopsy-confirmed cohort from China.The prevalence of CAA was high in the elderly Chinese sample.Age and apolipoprotein E (APOE) ε4 allele were related to the prevalence of CAA.CAA and Alzheimer's disease (AD) were frequently co-occurred and significantly associated.Subjects with both CAA and AD exhibited aggravated cognitive impairment.
Collapse
Affiliation(s)
- Xiang‐Sha Yin
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Jianru Sun
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Xue Wang
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Wei Wu
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Zhen Chen
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Di Zhang
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Yuanyuan Xu
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Yongmei Chen
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Wenying Qiu
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Xiaojing Qian
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
| | - Jun Ni
- Department of NeurologyState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Chao Ma
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterJoint Laboratory of Anesthesia and PainChinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
- National Human Brain Bank for Development and FunctionBeijingChina
- Chinese Institute for Brain ResearchBeijingChina
| |
Collapse
|
39
|
Bonnar O, Eyre B, van Veluw SJ. Perivascular brain clearance as a therapeutic target in cerebral amyloid angiopathy and Alzheimer's disease. Neurotherapeutics 2025; 22:e00535. [PMID: 39890534 DOI: 10.1016/j.neurot.2025.e00535] [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: 10/10/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025] Open
Abstract
Although distinct diseases, both cerebral amyloid angiopathy (CAA) and Alzheimer's disease (AD) are characterized by the aggregation and accumulation of amyloid-β (Aβ). This is thought to be due, in part, to impaired perivascular Aβ clearance from the brain. This shared failure in both diseases presents a common opportunity for therapeutic intervention. In this review we discuss the idea that promoting perivascular brain clearance could be an effective strategy for safely reducing Aβ levels in CAA and AD thereby improving clinical outcomes, most notably hemorrhagic stroke and cognitive decline. We will explore the evidence for the different forces that are thought to drive perivascular brain clearance, review the literature on potential strategies for potentiating these driving forces, and finally we will discuss the substantial translational challenges and considerations that would accompany such an intervention.
Collapse
Affiliation(s)
- Orla Bonnar
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Beth Eyre
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Susanne J van Veluw
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA; J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Zhang C, Tai Y, Kong M, Jia P, Ma G, Ba M. Synergistic associations of amyloid-β and phosphorylated tau with tau aggregation and cognitive decline in Alzheimer's disease. J Alzheimers Dis 2025; 104:1036-1044. [PMID: 40091557 DOI: 10.1177/13872877251322196] [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: 03/19/2025]
Abstract
BackgroundThe pathological hallmarks of Alzheimer's disease (AD) include the amyloid-β (Aβ) plaques and phosphorylated tau (p-tau) forming neurofibrillary tangles. Understanding the pathophysiological cascade related to Aβ and tau process is crucial.ObjectiveTo investigate the impact of Aβ positron emission tomography (PET) and cerebrospinal fluid (CSF) p-tau on tau pathology and cognitive decline in AD.MethodsWe analyzed 319 older individuals from the Alzheimer's Disease Neuroimaging Initiative (ADNI) who underwent Aβ (18F-florbetapir or 18F-florbetaben) and tau (18F-flortaucipir) PET scans, along with CSF and cognitive assessments. Aβ positivity (A+) was determined by global standardized uptake value ratio thresholds of ≥1.11 for 18F-florbetapir or ≥1.08 for 18F-florbetaben, while p-tau positivity (T+) was defined as CSF p-tau181 levels ≥23 pg/ml. Linear mixed regression models were used to assess the effects of PET Aβ and CSF p-tau181 levels on tau accumulation in predefined Braak regions and cognitive function over time.ResultsOur results revealed significant differences in PET tau pathology and cognitive decline between A + and A- individuals. We observed that interactions between Aβ and p-tau proteins were associated with tau accumulation and cognitive decline. Additionally, A-/T + individuals exhibited higher levels of tau accumulation in all Braak regions compared to A-/T- counterparts, suggesting a potential independent role of p-tau in tau pathology in the absence of Aβ.ConclusionsOur findings suggest that Aβ positivity and elevated CSF p-tau181 levels were associated with tau accumulation and cognitive decline, highlighting the relevance of soluble p-tau as a potential biomarker for further investigation.
Collapse
Affiliation(s)
- Chunhua Zhang
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
- Department of Neurology, Jiaozhou Branch of Shanghai East Hospital, Tongji University, Jiaozhou, China
| | - Yaojun Tai
- Department of Neurology, Jiaozhou Branch of Shanghai East Hospital, Tongji University, Jiaozhou, China
| | - Min Kong
- Department of Neurology, Yantaishan Hospital, Yantai, China
| | - Pengyuan Jia
- Department of Neurology, Jiaozhou Branch of Shanghai East Hospital, Tongji University, Jiaozhou, China
| | - Guozhao Ma
- Department of Neurology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Maowen Ba
- Department of Neurology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, China
| |
Collapse
|
41
|
Na D, Yang Y, Xie L, Piekna-Przybylska D, Bunn D, Shamambo M, White P. The Auditory Brainstem Response Diagnoses Alzheimer-Like Disease in the 5xFAD Mouse Model. eNeuro 2025; 12:ENEURO.0049-25.2025. [PMID: 40199587 PMCID: PMC12017886 DOI: 10.1523/eneuro.0049-25.2025] [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: 01/29/2025] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
Early and accurate diagnosis of Alzheimer's disease (AD) will be key for effective personalized treatment plans ( Cummings, 2023). Significant difficulties in auditory processing have been frequently reported in many patients with mild cognitive impairment, the prodromal form of AD ( Tarawneh et al., 2022), making it an outstanding candidate as AD diagnostic biomarker. However, the efficiency of diagnosis with this parameter has not been explored. Here we show that when male mice with amyloidosis begin to show memory decline, changes in the auditory brainstem response (ABR) to clicks enable the reliable diagnosis of disease using a machine learning algorithm. Interpretation of the machine learning diagnosis revealed that the upper levels of the auditory pathway, including the inferior colliculus, were the probable sources of the defects. Histological analyses show that in these locations, neuroinflammation and plaque deposition temporally correlate with behavioral changes consistent with memory loss. While these findings are tempered by the caveat that they derive from amyloidosis mice, we propose that ABR measurements be evaluated as an additional rapid, low-cost, noninvasive biomarker to assist the diagnostic testing of early-stage AD.
Collapse
Affiliation(s)
- Daxiang Na
- Department of Neuroscience, Ernst J. Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, New York 14642
| | - Yidan Yang
- School of Mathematical Sciences, Rochester Institute of Technology, Rochester, New York 14623
| | - Li Xie
- Department of Biomedical Genetics, University of Rochester, Rochester, New York 14642
| | - Dorota Piekna-Przybylska
- Department of Neuroscience, Ernst J. Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, New York 14642
| | - Dominic Bunn
- Department of Neuroscience, Ernst J. Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, New York 14642
| | - Maleelo Shamambo
- Department of Neuroscience, Ernst J. Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, New York 14642
| | - Patricia White
- Department of Neuroscience, Ernst J. Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, New York 14642
| |
Collapse
|
42
|
Jia L, Ke Y, Zhao S, Liu J, Luo X, Cao J, Liu Y, Guo Q, Chen WH, Chen F, Wang J, Wu H, Ding J, Zhao XM. Metagenomic analysis characterizes stage-specific gut microbiota in Alzheimer's disease. Mol Psychiatry 2025:10.1038/s41380-025-02973-7. [PMID: 40164697 DOI: 10.1038/s41380-025-02973-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 02/12/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with a decade-long preclinical pathological period that can be divided into several stages. Emerging evidence has revealed that the microbiota-gut-brain axis plays an important role in AD pathology. However, the role of gut microbiota in different AD stages has not been well characterized. In this study, we performed fecal shotgun metagenomic analysis on a Chinese cohort with 476 participants across five stages of AD pathology to characterize stage-specific alterations in gut microbiota and evaluate their diagnostic potential. We discovered extensive gut dysbiosis that is associated with neuroinflammation and neurotransmitter dysregulation, with over 10% of microbial species and gene families showing significant alterations during AD progression. Furthermore, we demonstrated that microbial gene families exhibited strong diagnostic capabilities, evidenced by an average AUC of 0.80 in cross-validation and 0.75 in independent external validation. In the optimal model, the most discriminant gene families are primarily involved in the metabolism of carbohydrates, amino acids, energy, glycan and vitamins. We found that stage-specific microbial gene families in AD pathology could be validated by an in vitro gut simulator and were associated with specific genera. We also observed that the gut microbiota could affect the progression of cognitive decline in 5xFAD mice through fecal microbiota transplantation, which could be used for early intervention of AD. Our multi-stage large cohort metagenomic analysis demonstrates that alterations in gut microbiota occur from the very early stages of AD pathology, offering important etiological and diagnostic insights.
Collapse
Affiliation(s)
- Longhao Jia
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China
| | - Yize Ke
- Fudan Microbiome Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, and Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shuo Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Jinxin Liu
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China
| | - Xiaohui Luo
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China
| | - Jixin Cao
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China
| | - Yujia Liu
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Wei-Hua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Institution of Medical Artificial Intelligence, Binzhou Medical University, Yantai, 264003, China
| | - Feng Chen
- Department of Radiology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, Hainan, 570311, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| | - Hao Wu
- Fudan Microbiome Center, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, and Center for Obesity and Hernia Surgery, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China.
| | - Xing-Ming Zhao
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China.
- Lingang Laboratory, Shanghai, 200031, China.
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
- MOE Key Laboratory of Computational Neuroscience and Brain‑Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China.
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang, 313000, China.
| |
Collapse
|
43
|
Kshirsagar S, Alvir RV, Pradeepkiran JA, Reddy AP, Reddy PH. Therapeutic potential of DDQ in enhancing mitochondrial health and cognitive function in Late-Onset Alzheimer's disease. Mitochondrion 2025; 83:102036. [PMID: 40158867 DOI: 10.1016/j.mito.2025.102036] [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: 12/09/2024] [Revised: 02/15/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline, mitochondrial dysfunction, and neuroinflammation. This study evaluates the therapeutic potential of DDQ, a small molecule in the humanized Abeta knockin (hAbKI) mice that represents late-onset AD. Our findings demonstrate that DDQ treatment significantly improves cognitive performance as assessed through behavioral tests, including the rotarod, open field, Y-maze, and Morris water maze, compared to untreated hAbKI mice. At the molecular level, DDQ promoted mitochondrial biogenesis, as evidenced by enhanced expression of key proteins like PGC1α, NRF1, and TFAM. Additionally, DDQ treatment facilitated mitophagy, as indicated by elevated levels of PINK1 and Parkin, and reduced neuroinflammation, reflected by decreased Iba1 and GFAP levels. Transmission electron microscopy analysis revealed a marked improvement in mitochondrial morphology, with increased mitochondrial length and reduced mitochondrial numbers in DDQ-treated mice. Furthermore, DDQ treatment led to an increase in mitophagic vacuoles, suggesting that it effectively removes dysfunctional mitochondria. Taken together, for the first time, our study results support the potential of DDQ as a promising neuroprotective agent for late-onset AD, addressing mitochondrial dysfunction, neuroinflammation, and cognitive decline. Our study focused on developing small molecules that modulate mitophagy, mitochondrial dynamics and neuroinflammatory pathways for aging, AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Rainier Vladlen Alvir
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | - Arubala P Reddy
- Department of Nutrition, Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
44
|
Zhai Y, Lu K, Yuan Y, Zhang Z, Xue L, Zhao F, Xu X, Wang H. Semaglutide improves cognitive function and neuroinflammation in APP/PS1 transgenic mice by activating AMPK and inhibiting TLR4/NF-κB pathway. J Alzheimers Dis 2025:13872877251329439. [PMID: 40151913 DOI: 10.1177/13872877251329439] [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: 03/29/2025]
Abstract
BackgroundAlzheimer's disease (AD) causes cognitive function disorder and has become the preeminent cause of dementia. Glucagon-like peptide-1 (GLP-1) receptor agonists, semaglutide, have shown positive effects on promoting the cognitive function. However, research about the mechanism of semaglutide as a therapeutic intervention in AD is sparse.ObjectiveThis study was to investigate the therapeutic efficacy of semaglutide in a transgenic mouse model of AD pathology and explored the detailed mechanism by semaglutide modulated neuroinflammatory processes.MethodsMale amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice were treated with semaglutide or vehicle for 8 weeks. Morris water maze test was used to assess the therapeutic efficacy of semaglutide on recognition function. Pathology analysis was performed to detect the deposition of amyloid plaques. High-throughput sequencing analysis was applied to specify the mechanism. Microglia and astrocyte activation were assessed with immunofluorescent staining. Inflammation cytokine levels were evaluated with enzyme-linked immunosorbent assay (ELISA). Related proteins and pathway were evaluated with western blot.ResultsSemaglutide treatment attenuated Aβ accumulation and enhanced cognitive function in APP/PS1 transgenic mice. Through transcriptomic profiling, immunohistochemical staining, and ELISA, semaglutide was substantiated to inhibit the overactivation of microglia and astrocytes, as well as to curtail the secretion of inflammatory mediators. Furthermore, semaglutide robustly activated AMP-activated protein kinase (AMPK) and suppressed the toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling cascade, thus reducing the Aβ deposition and dampening the inflammatory cascade.ConclusionsThe results demonstrated that semaglutide mitigated neuroinflammation and decelerated the advance of AD in APP/PS1 transgenic mice.
Collapse
Affiliation(s)
- Yanyu Zhai
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Kaili Lu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Yuan Yuan
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ziyao Zhang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Lixia Xue
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Fei Zhao
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Xiaofeng Xu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| | - Hongmei Wang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai 200233, China
| |
Collapse
|
45
|
Walsh AE, Lukens JR. Harnessing microglia-based cell therapies for the treatment of neurodegenerative diseases. Curr Opin Immunol 2025; 94:102552. [PMID: 40138748 DOI: 10.1016/j.coi.2025.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
Given the growing evidence linking microglia to the onset and progression of various neurodegenerative diseases, these brain-resident macrophages have emerged as a promising cell type for targeted therapeutic interventions. This review highlights recent studies that utilized innovative, microglia-focused strategies for the treatment of diverse neurodegenerative disorders including lysosomal storage disorders, granulin frontotemporal dementia, and Alzheimer's disease. Cutting-edge therapeutic strategies range from replacing faulty microglia with peripheral macrophage precursors or induced human pluripotent stem cell-derived microglia to engineering microglia that target toxic aggregates or deliver remediating payloads. We also examine the potential limitations as well as the clinical benefits of these strategies.
Collapse
Affiliation(s)
- Adeline E Walsh
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA; Molecular Physiology and Biological Physics Graduate Program, UVA, Charlottesville, VA 22908, USA; Biotechnology Training Program, UVA, Charlottesville, VA 22908, USA
| | - John R Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA; Molecular Physiology and Biological Physics Graduate Program, UVA, Charlottesville, VA 22908, USA; Biotechnology Training Program, UVA, Charlottesville, VA 22908, USA.
| |
Collapse
|
46
|
Liu MN, Chang HI, Huang SH, Huang CW, Hsu SW, Lin KJ, Ho TY, Huang KL, Cheng CM, Chang CC. Development and validation of global tau severity score in Alzheimer's disease using Florzolotau (18F) PET. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111343. [PMID: 40147808 DOI: 10.1016/j.pnpbp.2025.111343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Tau-specific positron emission tomography (tau-PET) is valuable for assessing Alzheimer's disease (AD) severity, with phenotypic differences between young-onset AD (YOAD) and late-onset AD (LOAD) likely driven by distinct relationships between tau pathology and cognition. OBJECTIVE This study developed a global tau severity (gTS) scale using Florzolotau (18F) PET and compared it with the CenTauR score for standardizing tau burden quantification. METHODS A total of 186 participants were enrolled, including a pilot group (15 cognitive unimpaired controls [CTL], 15 AD patients) and a validation group (27 CTL, 67 YOAD, and 62 LOAD patients). In the validation group, cutoffs for diagnosing YOAD and LOAD using the gTS or CenTauR score were calculated. RESULTS The white matter region was identified as the most suitable reference for Florzolotau (18F). The gTS cutoff values of 24.1 for both AD and YOAD and 34.1 for LOAD demonstrated the highest diagnostic accuracy, as indicated by the area under the curve (AUC). The gTS score showed a higher AUC compared to CenTauR in YOAD versus CTL or LOAD versus CTL. The gTS scores significantly predicted total scores and subdomains on cognitive ability screening instruments. Cognitive-gTS curve features were found to have quadratic and linear relationships with YOAD and LOAD, respectively, illustrating different relationships between gTS scores and cognition. CONCLUSION The gTS score, derived from Florzolotau (18F) PET scans, provides significant predictions regarding tau burden and cognitive measurements. The higher AUC of gTS compared to the CenTauR universal scores indicates that gTS scores offer a robust method for differentiating AD from CTL.
Collapse
Affiliation(s)
- Mu-N Liu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei 112201, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hsin-I Chang
- Department of Neurology, Cognition and Aging Center, Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shu-Hua Huang
- Department of Nuclear medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chi-Wei Huang
- Department of Neurology, Cognition and Aging Center, Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shih-Wei Hsu
- Department of Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear medicine, Lin-Kou Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Tsung-Ying Ho
- Department of Nuclear medicine, Lin-Kou Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Kuo-Lun Huang
- Department of Neurology, Lin-Kou Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Ming Cheng
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei 112201, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Chiung-Chih Chang
- Department of Neurology, Cognition and Aging Center, Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
47
|
Zhou Q, Wang W, Deng C. Advancements in Proteolysis Targeting Chimeras for Targeted Therapeutic Strategies in Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04838-0. [PMID: 40133753 DOI: 10.1007/s12035-025-04838-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/09/2025] [Indexed: 03/27/2025]
Abstract
The presence of hyperphosphorylated Tau proteins, which mislocalize and form neurofibrillary tangles, and the accumulation of amyloid-β plaques are hallmark features of Alzheimer's disease (AD). These toxic protein aggregates contribute to synaptic impairment and neuronal dysfunction, underscoring the need for strategies aimed at effectively clearing or reducing these aggregates in the treatment of AD. In recent years, proteolysis targeting chimera (PROTAC) technology has emerged as a promising approach for selectively degrading dysfunctional proteins rather than merely inhibiting their function. This approach holds great potential for developing more effective interventions that could slow AD progression and improve patient outcomes. In this review, we first examine the pathological mechanisms underlying AD, focusing on abnormal protein degradation and accumulation. We then explore the evolution of PROTAC technology, its mechanisms of action, and the current status of drug development. Finally, we discuss the latest findings regarding the application of PROTACs in AD therapy, highlighting the potential benefits and limitations of this technology. Although promising, further clinical research is necessary to fully assess the safety and efficacy of PROTAC-based therapies for AD treatment.
Collapse
Affiliation(s)
- Qiuzhi Zhou
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weixia Wang
- School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing, 100191, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
48
|
Ramesh M, Govindaraju T. MiR-7a-Klf4 axis as a regulator and therapeutic target of neuroinflammation and ferroptosis in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644978. [PMID: 40196619 PMCID: PMC11974717 DOI: 10.1101/2025.03.24.644978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Neuroinflammation and ferroptosis significantly contribute to neuronal death in Alzheimer's disease (AD) and other neurodegenerative disorders. MicroRNAs (miRNAs) are crucial regulators of these pathological processes. We employed transcriptomic analysis in an APP/PSEN1 Tg AD mouse model to identify dysregulated miRNAs and construct a miRNA-mRNA-pathway network. We discovered increased miR7a expression in the AD brain, targeting Krüppel-like factor 4 (Klf4), a transcriptional factor implicated in Aβ oligomer-induced neuroinflammation and RSL3-induced neuronal ferroptosis. Elevated Klf4 levels in AD mice brains suggest its involvement in AD pathology. The miR-7a mediated silencing of Klf4 alleviates neuroinflammation by modulating NF-κB, iNOS, and NLRP3 pathways, and inhibition of ferroptosis by targeting labile iron levels, GPX4, Nrf2 pathway, and mitochondrial damage. These findings highlight the neuroprotective role of miR-7a and its potential as RNA therapeutic. Pharmacological targeting of the miR-7a-Klf4 axis with blood-brain-barrier (BBB)-permeable compound effectively mitigates neuroinflammation and ferroptosis, suggesting the miR-7a-Klf4 axis as a novel therapeutic target for AD. GRAPHICAL ABSTRACT
Collapse
|
49
|
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 2025:13872877251326286. [PMID: 40105503 DOI: 10.1177/13872877251326286] [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: 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
|
50
|
Parhizkar S, Holtzman DM. The night's watch: Exploring how sleep protects against neurodegeneration. Neuron 2025; 113:817-837. [PMID: 40054454 PMCID: PMC11925672 DOI: 10.1016/j.neuron.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/15/2024] [Accepted: 02/04/2025] [Indexed: 03/21/2025]
Abstract
Sleep loss is often regarded as an early manifestation of neurodegenerative diseases given its common occurrence and link to cognitive dysfunction. However, the precise mechanisms by which sleep disturbances contribute to neurodegeneration are not fully understood, nor is it clear why some individuals are more susceptible to these effects than others. This review addresses critical unanswered questions in the field, including whether sleep disturbances precede or result from neurodegenerative diseases, the functional significance of sleep changes during the preclinical disease phase, and the potential role of sleep homeostasis as an adaptive mechanism enhancing resilience against cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Samira Parhizkar
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|