1
|
de Barros EC. Regulation of non-emotional memory through α 1-adrenergic receptors activation: A short review. IBRO Neurosci Rep 2025; 18:338-341. [PMID: 40034543 PMCID: PMC11872575 DOI: 10.1016/j.ibneur.2025.02.005] [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: 01/13/2025] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 03/05/2025] Open
Abstract
The α1 adrenergic receptors (α1-ARs) play a central role in the regulation of synaptic plasticity and memory, but their role in non-emotional memory is still poorly understood. This review summarizes recent advances in understanding the functions of α1-ARs and highlights their contributions to synaptic efficacy, long-term potentiation (LTP), and long-term depression (LTD) in the hippocampus and neocortex. There is evidence that α1-AR activation occurs through intracellular pathways such as Gq-protein signaling, MAPK, and cAMP cascades. Furthermore, α1-ARs are emerging as promising therapeutic targets in neurodegenerative diseases, including Alzheimer's disease (AD), due to their capability to modulate cognition and neuronal plasticity. New insights into positive allosteric modulators (PAMs) that cross the blood-brain barrier provide a potential avenue for safer and more effective therapies. This review highlights the need for further research to improve the understanding of α1-ARs and their potential for memory enhancement and neuroprotection.
Collapse
Affiliation(s)
- Eugénia Correia de Barros
- Department of Neurological Sciences and Mental Health, Faculty of Medicine, Agostinho Neto University, University of Lisbon, Lisboa, Portugal
| |
Collapse
|
2
|
Xie L, Sheehy RN, Muneer A, Xiong Y, Wrobel JA, Zhang F, Park KS, Velez J, Liu J, Luo YJ, Asrican B, Dong P, Li YD, Damian C, Quintanilla L, Li Y, Xu C, Deshmukh M, Coleman LG, Ming GL, Song H, Wen Z, Jin J, Song J, Chen X. Development of a brain-penetrant G9a methylase inhibitor to target Alzheimer's disease-associated proteopathology. Nat Commun 2025; 16:4222. [PMID: 40328756 PMCID: PMC12056044 DOI: 10.1038/s41467-025-59128-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Current Aβ-targeting therapeutics for Alzheimer's disease (AD) only slow cognitive decline due to poor understanding of AD pathogenesis. Here we describe a mechanism of AD pathogenesis in which the histone methyltransferase G9a noncanonically regulates translation of hippocampal proteins associated with AD pathology. Correspondingly, we developed a brain-penetrant inhibitor of G9a, MS1262, which restored both age-related learning & memory and noncognitive functions in multiple AD mouse models. Further, comparison of AD pathology-correlated mouse proteomes with those of AD patients found G9a regulates pathological pathways that promote Aβ and neurofibrillary tangles. This mouse-to-human overlap of G9a regulated AD-associated pathologic proteins supports at the molecular level the efficacy of targeting G9a translational mechanism for treating AD patients. Additionally, MS1262 treatment reversed the AD-characteristic expression or phosphorylation of multiple clinically validated biomarkers of AD that have the potential to be used for early-stage AD diagnosis and companion diagnosis of individualized drug effects.
Collapse
Affiliation(s)
- Ling Xie
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ryan N Sheehy
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adil Muneer
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John A Wrobel
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Kwang-Su Park
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia Velez
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yan-Jia Luo
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brent Asrican
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ping Dong
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ya-Dong Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Corina Damian
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Luis Quintanilla
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yongyi Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chongchong Xu
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Xian Chen
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Vigers MP, Lobo S, Najafi S, Dubose A, Tsay K, Ganguly P, Longhini AP, Jin Y, Buratto SK, Kosik KS, Shell MS, Shea JE, Han S. Water-directed pinning is key to tau prion formation. Proc Natl Acad Sci U S A 2025; 122:e2421391122. [PMID: 40294272 DOI: 10.1073/pnas.2421391122] [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: 10/24/2024] [Accepted: 03/10/2025] [Indexed: 04/30/2025] Open
Abstract
Tau forms fibrillar aggregates that are pathological hallmarks of a family of neurodegenerative diseases known as tauopathies. The synthetic replication of disease-specific fibril structures is a critical gap for developing diagnostic and therapeutic tools. This study debuts a strategy of identifying a critical and minimal folding motif in fibrils characteristic of tauopathies and generating seeding-competent fibrils from the isolated tau peptides. The 19-residue jR2R3 peptide (295 to 313) which spans the R2/R3 splice junction of tau, and includes the P301L mutation, is one such peptide that forms prion-competent fibrils. This tau fragment contains the hydrophobic VQIVYK hexapeptide that is part of the core of all known pathological tau fibril structures and an intramolecular counterstrand that stabilizes the strand-loop-strand (SLS) motif observed in 4R tauopathy fibrils. This study shows that P301L exhibits a duality of effects: it lowers the barrier for the peptide to adopt aggregation-prone conformations and enhances the local structuring of water around the mutation site to facilitate site-directed pinning and dewetting around sites 300-301 to achieve in-register stacking of tau to cross β-sheets. We solved a 3 Å cryo-EM structure of jR2R3-P301L fibrils in which each protofilament layer contains two jR2R3-P301L copies, of which one adopts a SLS fold found in 4R tauopathies and the other wraps around the SLS fold to stabilize it, reminiscent of the three- and fourfold structures observed in 4R tauopathies. These jR2R3-P301L fibrils are competent to template full-length 4R tau in a prion-like manner.
Collapse
Affiliation(s)
- Michael P Vigers
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106
| | - Samuel Lobo
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106
| | - Saeed Najafi
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106
| | - Austin Dubose
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106
| | - Karen Tsay
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106
| | - Pritam Ganguly
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106
- Department of Physics, University of California, Santa Barbara, CA 93106
| | - Andrew P Longhini
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106
| | - Yingying Jin
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106
| | - Steven K Buratto
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106
| | - Kenneth S Kosik
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106
| | - M Scott Shell
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106
| | - Joan-Emma Shea
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106
- Department of Physics, University of California, Santa Barbara, CA 93106
| | - Songi Han
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106
- Department of Chemistry, Northwestern University, Evanston, IL 60208
| |
Collapse
|
4
|
Wu Y, Wei M, Wang M, Guo M, Yu H, Chen Y, Xu T, Zhou Y. Schisandra total lignans ameliorate neuronal ferroptosis in 3xTg-AD mice via regulating NADK/NADPH/GSH pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156612. [PMID: 40088743 DOI: 10.1016/j.phymed.2025.156612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/22/2024] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder with limited treatments. Schisandra total lignans (STL), the primary active component of Schisandra chinensis, shows potential in alleviating AD-related symptoms, though the mechanisms remain unclear. PURPOSE Considering the promoting effect of neuronal ferroptosis on AD and the neuroprotective activity of STL, this study aimed to investigate the impact of STL on AD neuronal ferroptosis and elucidate its underlying mechanisms. METHODS This study used 3xTg-AD mice and SH-SY5Y cells overexpressing APPswe as models. UHPLC/Q-TOF-MS was applied for identifying components in STL extract and the plasma of 3xTg-AD mice, as well as to detect cellular endogenous metabolites for one-carbon metabolism analysis. Behavioral tests, including the Y maze, novel object recognition, Morris water maze, and open field, were conducted to assess the cognitive function and emotional state. Histopathological examinations were performed using immunofluorescence, immunohistochemistry, Nissl staining, and transmission electron microscopy. The GSH, GSSG, NAD(H), NADP(H), and MDA levels, as well as GPX and GR activity were measured using assay kits. ROS, Fe2+, and lipid peroxidation levels were detected with probes. Protein expression was evaluated by Western blot. Molecular docking, molecular dynamics simulations and cellular thermal shift assay were performed to analyze the STL-NADK interactions. RESULTS Behavioral tests indicated that STL alleviated cognitive impairments and anxiety in 3xTg-AD mice. Histological analysis showed that STL decreased hippocampal Aβ levels, inhibited hippocampal neuronal ferroptosis, and mitigated synaptic damage. Cellular assays demonstrated that STL alleviated APPswe overexpression-induced ferroptosis and synaptic damage by activating the NADK/NADPH/GSH pathway, with NADK knockdown abolishing this neuroprotective effect of STL. Computational analysis and cellular thermal shift assay identified Gomisin D as the key STL component with strong affinity for NADK, driving its neuroprotective effects. CONCLUSION NADK emerges as a novel potential therapeutic target for AD, with STL activating NADK, promoting NADPH and GSH production, thereby mitigating neuronal ferroptosis in AD.
Collapse
Affiliation(s)
- Yuying Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Mengying Wei
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Mengyao Wang
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Minsong Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hengyuan Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yong Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Tengfei Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Yuan Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
5
|
Tenner AJ, Petrisko TJ. Knowing the enemy: strategic targeting of complement to treat Alzheimer disease. Nat Rev Neurol 2025; 21:250-264. [PMID: 40128350 DOI: 10.1038/s41582-025-01073-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2025] [Indexed: 03/26/2025]
Abstract
The complement system protects against infection, positively responds to tissue damage, clears cell debris, directs and modulates the adaptive immune system, and functions in neuronal development, normal synapse elimination and intracellular metabolism. However, complement also has a role in aberrant synaptic pruning and neuroinflammation - processes that lead to a feedforward loop of inflammation, injury and neuronal death that can contribute to neurodegenerative and neurological disorders, including Alzheimer disease. This Review provides justification, largely from preclinical mouse models but also from correlates with human tissue and biomarkers, for targeting specific complement components for therapeutic intervention in Alzheimer disease. We discuss promising strategies to slow the progression of cognitive loss with minimal undesired effects. The diverse interactions and functions of complement system components can influence biological processes in the healthy and diseased brain; here, these functions are described as a prerequisite to selecting appropriate, safe and effective therapeutic targets for translation to the clinic.
Collapse
Affiliation(s)
- Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA.
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA.
| | - Tiffany J Petrisko
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
6
|
Korkmaz F, Sims S, Sen F, Sultana F, Laurencin V, Cullen L, Pallapati A, Liu A, Chen R, Rojekar S, Pevnev G, Cheliadinova U, Vasilyeva D, Burganova G, Macdonald A, Saxena M, Goosens K, Rosen CJ, Barak O, Lizneva D, Gumerova A, Ye K, Ryu V, Yuen T, Frolinger T, Zaidi M. Gene-dose-dependent reduction of Fshr expression improves spatial memory deficits in Alzheimer's mice. Mol Psychiatry 2025; 30:2119-2126. [PMID: 39548323 DOI: 10.1038/s41380-024-02824-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
High post-menopausal levels of the pituitary gonadotropin follicle-stimulating hormone (FSH) are strongly associated with the onset of Alzheimer's disease (AD). We have shown recently that FSH directly activates the hippocampal FSH receptors (FSHRs) to drive AD-like pathology and memory loss in mice. To unequivocally establish a role for FSH in memory loss, we depleted the Fshr on a 3xTg background and utilized Morris Water Maze to study deficits in spatial memory. 3xTg;Fshr+/+ mice displayed impaired spatial memory at 5 months of age. The loss of memory acquisition and retrieval were both rescued in 3xTg;Fshr-/- mice and, to a lesser extent, in 3xTg;Fshr+/- mice-documenting clear gene-dose-dependent prevention of spatial memory loss. Furthermore, at 5 and 8 months, sham-operated 3xTg;Fshr-/- mice showed better memory performance during the learning and/or retrieval phases, further suggesting that Fshr deletion prevents age-related progression of memory deficits. This prevention was not seen when mice were ovariectomized, except in the 8-month-old 3xTg;Fshr-/- mice. There was also a gene-dose-dependent reduction mainly in the amyloid β40 isoform in whole brain extracts. Finally, serum FSH levels <8 ng/mL in 16-month-old APP/PS1 mice were associated with better retrieval of spatial memory. Collectively, the data provide compelling genetic evidence for a protective effect of inhibiting FSH signaling on the progression of spatial memory deficits in mice and lay a firm foundation for the use of an FSH-blocking agent for the early prevention of memory loss in post-menopausal women.
Collapse
Affiliation(s)
- Funda Korkmaz
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven Sims
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fazilet Sen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Farhath Sultana
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Victoria Laurencin
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Liam Cullen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anusha Pallapati
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Avi Liu
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald Chen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Satish Rojekar
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgii Pevnev
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Uliana Cheliadinova
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Darya Vasilyeva
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guzel Burganova
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anne Macdonald
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mansi Saxena
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ki Goosens
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Orly Barak
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daria Lizneva
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anisa Gumerova
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, University of Chinese Academy of Science, Shenzhen, Guangdong, China
| | - Vitaly Ryu
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tony Yuen
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Tal Frolinger
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Mone Zaidi
- Mount Sinai Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
7
|
He J, Sun S, Wang H, Ying Z, Tam KY. Triple-Target Inhibition of Cholinesterase, Amyloid Aggregation, and GSK3β to Ameliorate Cognitive Deficits and Neuropathology in the Triple-Transgenic Mouse Model of Alzheimer's Disease. Neurosci Bull 2025; 41:821-836. [PMID: 39907971 PMCID: PMC12014999 DOI: 10.1007/s12264-025-01354-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/06/2024] [Indexed: 02/06/2025] Open
Abstract
Alzheimer's disease (AD) poses one of the most urgent medical challenges in the 21st century as it affects millions of people. Unfortunately, the etiopathogenesis of AD is not yet fully understood and the current pharmacotherapy options are somewhat limited. Here, we report a novel inhibitor, Compound 44, for targeting cholinesterases, amyloid-β (Aβ) aggregation, and glycogen synthase kinase 3β (GSK-3β) simultaneously with the aim of achieving symptomatic relief and disease modification in AD therapy. We found that Compound 44 had good inhibitory effects on all intended targets with IC50s of submicromolar or better, significant neuroprotective effects in cell models, and beneficial improvement of cognitive deficits in the triple transgenic AD (3 × Tg AD) mouse model. Moreover, we showed that Compound 44 acts as an autophagy regulator by inducing nuclear translocation of transcription factor EB through GSK-3β inhibition, enhancing the biogenesis of lysosomes and elevating autophagic flux, thus ameliorating the amyloid burden and tauopathy, as well as mitigating the disease phenotype. Our results suggest that triple-target inhibition via Compound 44 could be a promising strategy that may lead to the development of effective therapeutic approaches for AD.
Collapse
Affiliation(s)
- Junqiu He
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Shan Sun
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215127, China
| | - Hongfeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215127, China.
| | - Zheng Ying
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215127, China.
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
8
|
Billard JM, Ploux E, Largilliere S, Corvaisier S, Gorisse-Hussonnois L, Radzishevsky I, Wolosker H, Freret T. Early involvement of D-serine in β-amyloid-dependent pathophysiology. Cell Mol Life Sci 2025; 82:179. [PMID: 40293541 PMCID: PMC12037454 DOI: 10.1007/s00018-025-05691-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/04/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025]
Abstract
The N-methyl-D-aspartate subtype of glutamate receptors (NMDAR) is a key regulator of brain plasticity encoding learning and memory. In addition to glutamate, NMDAR activation requires the binding of the co-agonist D-serine. The beta-amyloid (Aß) peptide which accumulates in Alzheimer's disease (AD), affects the D-serine-dependent NMDAR activation in vitro, but whether this alteration would significantly contribute to AD-related pathophysiology and memory deficits remains unclear. Herein, we report a decrease in the maximal pool of recruitable NMDAR and in the expression of NMDAR-dependent long-term potentiation together with impaired basal neurotransmission at CA3/CA1 synapses from hippocampal slices of 5xFAD mouse, an AD-related model with elevated Aß levels. The NMDAR synaptic impairments develop from 1.5 to 2 months of age with the initial rise of Aß and is correlated to a transient increase in D-serine levels. Deficits in working and spatial memories as well as cognitive flexibility then occurred in 10-12 months-old animals. Importantly, the NMDA-related synaptic deregulations (but not the altered basal neurotransmission) and behavioral impairments (working and cognitive flexibility) are prevented or reduced (spatial memory) in 5xFAD mice devoid of D-serine after genetic deletion of its synthesis enzyme serine racemase. Altogether, these results therefore provide in vivo evidence for the implication of D-serine at least in the early pathogenic signatures of AD driven by the increase in amyloid load suggesting that the recent proposal of preventive therapy of AD by administration of the precursor L-serine remains questionable.
Collapse
Affiliation(s)
- J-M Billard
- Normandie University, UNICAEN, INSERM, GIP CYCERON, COMETE, Caen, France.
- UNICAEN COMETE, INSERM UMR S-1075, GIP CYCERON, Bat GMPc, Campus Horowitz, Bd Henri Becquerel, Caen, CS14032, France.
| | - E Ploux
- Normandie University, UNICAEN, INSERM, GIP CYCERON, COMETE, Caen, France
| | - S Largilliere
- Normandie University, UNICAEN, INSERM, GIP CYCERON, COMETE, Caen, France
| | - S Corvaisier
- Normandie University, UNICAEN, INSERM, GIP CYCERON, COMETE, Caen, France
| | | | - I Radzishevsky
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - H Wolosker
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - T Freret
- Normandie University, UNICAEN, INSERM, GIP CYCERON, COMETE, Caen, France.
| |
Collapse
|
9
|
Wang N, Li XZ, Jiang XW, Ning XY, Zhou LJ, Liu WJ, Wu Q, Wang XP, Xing Y, Qi ZT, Xu ZH, Song HT, Zhao QC. Multi-Target Inhibitor of ZJQ- 3 F Against AChE/BACE1/GSK3β Targets Improves the Cognitive Impairment of APP/PS1/Tau Triple-Transgenic Mouse Models of Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04982-7. [PMID: 40285940 DOI: 10.1007/s12035-025-04982-7] [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: 12/06/2023] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a multifactorial neuropathology characterized by the accumulation of amyloid-beta (Aβ) plaques, neurofibrillary tangles (NFTs) and cholinergic system dysfunction. At present, there is no effective treatment strategy for AD. Our previous research showed that ZJQ-3F acts as an inhibitor of AChE/BACE1/GSK3β, and showed good blood-brain barrier permeability, appropriate bioavailability and oral safety. In order to further study, the protective effect of ZJQ-3F on APP/PS1/Tau transgenic mice was determined. METHODS APP/PS1/Tau transgenic mice model of AD was treated with ZJQ-3F from the age of 8 to 12 months, and then behavioral tests was conducted. Western blot, immunohistochemistry and immunofluorescence staining were used to evaluate the level of tau protein, Aβ plaques and synaptic function. RESULTS Our results revealed that administration of ZJQ-3F could improve the cognitive function of APP/PS1/Tau transgenic mice. In addition, compared with APP/PS1/Tau mice, the protein expression levels of tau protein phosphorylation site at Ser396, Thr212 and Thr181 in the cortex and hippocampus of ZJQ-3F treated mice was significantly decreased. Moreover, the results showed that ZJQ-3F significantly reduced the deposition of Aβ in the cortex and hippocampus. Furthermore, the results indicated that the protein expression levels of PSD95, SYP and SYT in the cortex and hippocampus were increased markedly after ZJQ-3F was given. CONCLUSIONS Our studies suggest that the chronic administration of ZJQ-3F can improve learning and memory ability, reduce tau protein phosphorylation, reduce Aβ deposition and improve synaptic dysfunction in APP/PS1/Tau transgenic model of AD, indicating that ZJQ-3F can be used as a multi-target inhibitor to slow down the progress of AD.
Collapse
Affiliation(s)
- Nan Wang
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China
| | - Xin-Zhu Li
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Xiao-Wen Jiang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Xin-Yue Ning
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Li-Jun Zhou
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Wen-Jie Liu
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Qiong Wu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China
| | - Xin-Peng Wang
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Yu Xing
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Zhen-Tong Qi
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Zi-Hua Xu
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China
| | - Hong-Tao Song
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China.
- Department of Pharmacy, 900 Hospital of the Joint Logistics Team, Fuzhou, 350025, People's Republic of China.
| | - Qing-Chun Zhao
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China.
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China.
| |
Collapse
|
10
|
Luo M, Zhou J, Sun C, Chen W, Fu C, Si C, Zhang Y, Geng Y, Chen Y. APP β-CTF triggers cell-autonomous synaptic toxicity independent of Aβ. eLife 2025; 13:RP100968. [PMID: 40266681 PMCID: PMC12017768 DOI: 10.7554/elife.100968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025] Open
Abstract
Aβ is believed to play a significant role in synaptic degeneration observed in Alzheimer's disease and is primarily investigated as a secreted peptide. However, the contribution of intracellular Aβ or other cleavage products of its precursor protein (APP) to synaptic loss remains uncertain. In this study, we conducted a systematic examination of their cell-autonomous impact using a sparse expression system in rat hippocampal slice culture. Here, these proteins/peptides were overexpressed in a single neuron, surrounded by thousands of untransfected neurons. Surprisingly, we found that APP induced dendritic spine loss only when co-expressed with BACE1. This effect was mediated by β-CTF, a β-cleavage product of APP, through an endosome-related pathway independent of Aβ. Neuronal expression of β-CTF in mouse brains resulted in defective synaptic transmission and cognitive impairments, even in the absence of amyloid plaques. These findings unveil a β-CTF-initiated mechanism driving synaptic toxicity irrespective of amyloid plaque formation and suggest a potential intervention by inhibiting the endosomal GTPase Rab5.
Collapse
Affiliation(s)
- Menguxn Luo
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jia Zhou
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Cailu Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Wanjia Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Chaoying Fu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of SciencesShanghaiChina
| | - Chenfang Si
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yaoyang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of SciencesShanghaiChina
| | - Yang Geng
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of SciencesShanghaiChina
| | - Yelin Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of SciencesShanghaiChina
| |
Collapse
|
11
|
Vieira A, Abatti M, Michels M, Goulart A, Faller CJ, Borges H, Fernandes F, Dominguini D, Rocha L, Córneo E, Dias R, Dal-Pizzol F. The Impact of Biological Sex And High-Fat High-Fructose Diet on Brain Dysfunction in an Animal Model of Sepsis. Mol Neurobiol 2025:10.1007/s12035-025-04937-y. [PMID: 40268828 DOI: 10.1007/s12035-025-04937-y] [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: 01/02/2024] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
The aim of this study was to evaluate long-term inflammatory, biochemical and behavioral parameters in adult male and female Wistar rats submitted to a model of high-fat and high fructose diet and sepsis. In the study we used 8-month-old male and female rats. High-fat and high fructose diet was provided for 4 months, and sepsis was induced shortly afterwards. Behavioral tests were performed at 10, 30 and 60 days after sepsis induction, at 30- and 60-days metabolic parameters, leptin and cytokines (prefrontal cortex and hippocampus) were determined. High-fat and high-fructose diet was able to induce glucose intolerance. Sepsis favored anxious behavior at 10 days after sepsis, remaining at 30 days and with apparent improvement at 60 days in females and maintenance of behavior in males. Cognitive damage was observed both at 30 and 60 days in animals from both groups. Plasma metabolic parameters were elevated only males exposed to a high-fat high-fructose diet and submitted to CLP only at 30 days. Long-term brain inflammation was not consistently affected both by sex and high-fat and high fructose diet.The relationship between high-fat and high fructose diet, gender and sepsis is still contradictory, as are the mechanisms involved in this paradox. Models and analyses need to be standardized in order to better understand how this event occurs.
Collapse
Affiliation(s)
- Andriele Vieira
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil.
- UNESC - Universidade do Extremo Sul Catarinense, PPGCS - Programa de Pós-graduação em Ciências da Saúde, Endres: Av. Universitária, Bairro Universitário, Criciúma, SC, 1105, Brazil.
| | - Mariane Abatti
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Monique Michels
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Amanda Goulart
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Cristiano Julio Faller
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Heloisa Borges
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Filipe Fernandes
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Diogo Dominguini
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Luana Rocha
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Emily Córneo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Rodrigo Dias
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| |
Collapse
|
12
|
Boschen SL, A Mukerjee A, H Faroqi A, E Rabichow B, Fryer J. Research models to study lewy body dementia. Mol Neurodegener 2025; 20:46. [PMID: 40269912 PMCID: PMC12020038 DOI: 10.1186/s13024-025-00837-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Lewy body dementia (LBD) encompasses neurodegenerative dementias characterized by cognitive fluctuations, visual hallucinations, and parkinsonism. Clinical differentiation of LBD from Alzheimer's disease (AD) remains complex due to symptom overlap, yet approximately 25% of dementia cases are diagnosed as LBD postmortem, primarily identified by the presence of α-synuclein aggregates, tau tangles, and amyloid plaques. These pathological features position LBD as a comorbid condition of both Parkinson's disease (PD) and AD, with over 50% of LBD cases exhibiting co-pathologies. LBD's mixed pathology complicates the development of comprehensive models that reflect the full spectrum of LBD's etiological, clinical, and pathological features. While existing animal and cellular models have facilitated significant discoveries in PD and AD research, they lack specificity in capturing LBD's unique pathogenic mechanisms, limiting the exploration of therapeutic avenues for LBD specifically. This review assesses widely used PD and AD models in terms of their relevance to LBD, particularly focusing on their ability to replicate human disease pathology and assess treatment efficacy. Furthermore, we discuss potential modifications to these models to advance the understanding of LBD mechanisms and propose innovative research directions aimed at developing models with enhanced etiological, face, predictive, and construct validity.
Collapse
Affiliation(s)
- Suelen Lucio Boschen
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
- Department of Neurosurgery, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
| | - Aarushi A Mukerjee
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Ayman H Faroqi
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Ben E Rabichow
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - John Fryer
- Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ, 850054, USA
| |
Collapse
|
13
|
Johnson KJ, Johnson K, Grant A, Taglialatela G, Micci MA. Photobiomodulation therapy increases neural stem cell pool in aged 3xTg-AD mice. PLoS One 2025; 20:e0321668. [PMID: 40261888 PMCID: PMC12013953 DOI: 10.1371/journal.pone.0321668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/10/2025] [Indexed: 04/24/2025] Open
Abstract
Presently approved Alzheimer's Disease (AD) therapeutics are designed for targeted removal of the AD-related toxic protein aggregate amyloid-β (Aβ) and have only shown moderate efficacy at slowing disease progression. Reversal of cognitive decline requires both removal of toxic aggregates and repair of the cellular systems damaged by decades of exposure to these aggregates. Adult hippocampal neurogenesis (AHN) is one such system that is known to be affected early and severely in the development of AD. Moreover, preserved AHN is associated with cognitive resilience to AD neuropathology. Therefore, targeted therapies to improve or enhance neurogenesis should be considered in addition to the removal of toxic protein aggregates. Photobiomodulation (PBM) using 670 nm LED light has been shown to induce synaptic resilience to and removal of AD-related toxic protein aggregates. In this study, we aimed to assess the effect of PBM on a mouse model of advanced AD neuropathology. Transgenic 3xTg-AD mice (15- to 17-month old) were randomized to receive PBM or SHAM therapy for one month, followed by neuropathological assessments. Our results show that one month of PBM therapy reduces hyperphosphorylated tau burden and partially rescues AHN in aged 3xTg-AD mice as compared to SHAM-treated transgenic mice. These data support the notion that PBM has the potential to be an effective non-invasive therapy to help preserve AHN and reduce cognitive dysfunction in moderate to advanced AD.
Collapse
Affiliation(s)
- Kevin J. Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Neurobiology, Neuroscience Graduate Program, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kathia Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Auston Grant
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Giulio Taglialatela
- The Mitchell Center for Neurodegenerative Disorders, Department of Neurology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Maria-Adelaide Micci
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
14
|
Saaoud F, Liu L, Xu K, Lu Y, Shao Y, Ben Issa M, Jiang X, Wang X, Liu X, Autieri M, Wu S, Wei J, Yu J, Bouchareb R, Gillespie A, Luo JJ, Martinez L, Vazquez-Padron R, Sun J, Zhao H, Wang H, Pratico D, Yang X. Alzheimer's disease as an auto-innate immune pathology with potential cell trans-differentiation and enhanced trained immunity in 3xTg-AD mouse model. J Alzheimers Dis 2025:13872877251329583. [PMID: 40232249 DOI: 10.1177/13872877251329583] [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 neurodegenerative disorder characterized by memory impairment. Neuroinflammatory processes, mediated by glial and immune cells, contribute to neuronal damage. Emerging evidence implicates innate immune mechanisms, including trained immunity and cell trans-differentiation, in AD pathogenesis, though their roles remain unclear.ObjectiveTo investigate transcriptomic changes in the 3xTg-AD mouse model, focusing on trained immunity and cell trans-differentiation in disease mechanisms.MethodsRNA-sequencing was performed on brain tissue (cortex plus hippocampus) from 11-month-old female 3xTg-AD and wild-type mice (n = 3/group). Differentially expressed genes (fold change > 1.5, p < 0.05) were identified and followed by bioinformatics and knowledge-based transcriptomic profiling. Public AD datasets were also analyzed.Results3xTg-AD mice exhibited 316 upregulated and 412 downregulated genes. Downregulated genes included those for blood-brain barrier protein, while upregulated genes related to cerebrospinal fluid. Increased expression of proinflammatory markers, as well as genes related to cell differentiation, proliferation, activation, and adhesion. Upregulation of genes associated with cell migration and trans-differentiation suggests a potential role for inflammation and cellular plasticity. Additionally, genes involved in inflammasome pathways, immunometabolism, and trained immunity were upregulated. Mechanistically, these genes were modulated by knockdown of trained immunity promoter SET-7, overexpression of trained immunity inhibitor IL-37, and knockout of inflammasome genes IL-1 receptor, caspase-1, and pattern recognition receptor CD36.ConclusionsThe finding underscore the potential role of trained immunity and cell trans-differentiation in AD, revealing a mechanistic framework in which danger-associated molecular patterns drive innate immune responses, inflammasome activation, and cell plasticity contribute to AD, offering therapeutic targets for neuroinflammation and cellular reprograming.
Collapse
Affiliation(s)
- Fatma Saaoud
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Lu Liu
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Keman Xu
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Yifan Lu
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Ying Shao
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Mohammed Ben Issa
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Xiaohua Jiang
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xianwei Wang
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xiaolei Liu
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Michael Autieri
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
| | - Sheng Wu
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Juncheng Wei
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jun Yu
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Rihab Bouchareb
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Avrum Gillespie
- Section of Nephrology, Hypertension, and Kidney Transplantation, Department of Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jin Jun Luo
- Department of Neurology, Temple University, Philadelphia, PA, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Roberto Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jianxin Sun
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Huaqing Zhao
- Department of Biomedical Education and Data Sciences, Temple University, Philadelphia, PA, USA
| | - Hong Wang
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Domenico Pratico
- Alzheimer's Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xiaofeng Yang
- Lemole Center for Integrated Lymphatics and Vascular Research, Temple University, Philadelphia, PA, USA
- Lemole Center for Integrated Lymphatics and Vascular Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
15
|
Fourriere L, Gleeson PA. Organelle perturbation in Alzheimer's disease: do intracellular amyloid-ß and the fragmented Golgi mediate early intracellular neurotoxicity? Front Cell Dev Biol 2025; 13:1550211. [PMID: 40302938 PMCID: PMC12037564 DOI: 10.3389/fcell.2025.1550211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
Alzheimer's disease is a devastating and incurable neurological disease. Most of the current research has focused on developing drugs to clear the extracellular amyloid plaques in the brain of Alzheimer's disease patients. However, this approach is limited as it does not treat the underlying cause of the disease. In this review, we highlight the evidence in the field showing that the accumulation of intracellular toxic amyloid-ß could underpin very early events in neuronal death in both familial early-onset and sporadic late-onset alzheimer's disease. Indeed, intracellular amyloid-ß, which is produced within intracellular compartments, has been shown to perturb endosomal and secretory organelles, in different neuronal models, and the brain of Alzheimer's patients, leading to membrane trafficking defects and perturbation of neuronal function associated with cognition defects. The Golgi apparatus is a central transport and signaling hub at the crossroads of the secretory and endocytic pathways and perturbation of the Golgi ribbon structure is a hallmark of Alzheimer's disease. Here, we discuss the role of the Golgi as a major player in the regulation of amyloid-β production and propose that the Golgi apparatus plays a key role in a cellular network which can seed the onset of Alzheimer's disease. Moreover, we propose that the Golgi is central in an intracellular feedback loop leading to an enhanced level of amyloid-β production resulting in early neuronal defects before the appearance of clinical symptoms. Further advances in defining the molecular pathways of this intracellular feedback loop could support the design of new therapeutic strategies to target a primary source of neuronal toxicity in this disease.
Collapse
|
16
|
Dias OFM, Valle NME, Mamani JB, Costa CJS, Alves AH, Oliveira FA, Rego GNA, Galanciak MCS, Felix K, Nucci MP, Gamarra LF. Longitudinal Evaluation of the Detection Potential of Serum Oligoelements Cu, Se and Zn for the Diagnosis of Alzheimer's Disease in the 3xTg-AD Animal Model. Int J Mol Sci 2025; 26:3657. [PMID: 40332200 PMCID: PMC12026877 DOI: 10.3390/ijms26083657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of β-amyloid (Aβ) and hyperphosphorylated tau, leading to neuroinflammation, oxidative stress, and neuronal death. Early detection of AD remains a challenge, as clinical manifestations only emerge in the advanced stages, limiting therapeutic interventions. Minimally invasive biomarkers are essential for early identification and monitoring of disease progression. This study aims to evaluate the sensitivity of the relationship between serum oligoelement levels as biomarkers and the monitoring of AD progression in the 3xTg-AD model. Transgenic 3xTg-AD mice and C57BL/6 controls were evaluated over 12 months through serum oligoelement quantification using inductively coupled plasma mass spectrometry (ICP-MS), Aβ deposition via immunohistochemistry, and cognitive assessments using memory tests (Morris water maze and novel object recognition test), as well as spontaneous locomotion analysis using the open field test. The results demonstrated that oligoelements (copper, zinc, and selenium) were sensitive in detecting alterations in the AD group, preceding cognitive and motor deficits. Immunohistochemistry was performed for qualitative purposes, confirming the presence of β-amyloid in the CNS of transgenic animals. Up to the third month, labeling was moderate and restricted to neuronal cell bodies; from the fifth month onward, evident extracellular deposits emerged. Behavioral assessment indicated impairments in spatial and episodic memory, as well as altered locomotor patterns in AD mice. These findings reinforce that oligoelement variations may be associated with neurodegenerative processes, including oxidative stress and synaptic dysfunction. Thus, oligoelement analysis emerges as a promising approach for the early diagnosis of AD and the monitoring of disease progression, potentially contributing to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Olivia F. M. Dias
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (O.F.M.D.); (N.M.E.V.); (J.B.M.); (C.J.S.C.); (A.H.A.); (F.A.O.); (G.N.A.R.); (M.C.S.G.); (K.F.)
| | - Nicole M. E. Valle
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (O.F.M.D.); (N.M.E.V.); (J.B.M.); (C.J.S.C.); (A.H.A.); (F.A.O.); (G.N.A.R.); (M.C.S.G.); (K.F.)
| | - Javier B. Mamani
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (O.F.M.D.); (N.M.E.V.); (J.B.M.); (C.J.S.C.); (A.H.A.); (F.A.O.); (G.N.A.R.); (M.C.S.G.); (K.F.)
| | - Cicero J. S. Costa
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (O.F.M.D.); (N.M.E.V.); (J.B.M.); (C.J.S.C.); (A.H.A.); (F.A.O.); (G.N.A.R.); (M.C.S.G.); (K.F.)
| | - Arielly H. Alves
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (O.F.M.D.); (N.M.E.V.); (J.B.M.); (C.J.S.C.); (A.H.A.); (F.A.O.); (G.N.A.R.); (M.C.S.G.); (K.F.)
| | - Fernando A. Oliveira
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (O.F.M.D.); (N.M.E.V.); (J.B.M.); (C.J.S.C.); (A.H.A.); (F.A.O.); (G.N.A.R.); (M.C.S.G.); (K.F.)
| | - Gabriel N. A. Rego
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (O.F.M.D.); (N.M.E.V.); (J.B.M.); (C.J.S.C.); (A.H.A.); (F.A.O.); (G.N.A.R.); (M.C.S.G.); (K.F.)
| | - Marta C. S. Galanciak
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (O.F.M.D.); (N.M.E.V.); (J.B.M.); (C.J.S.C.); (A.H.A.); (F.A.O.); (G.N.A.R.); (M.C.S.G.); (K.F.)
| | - Keithy Felix
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (O.F.M.D.); (N.M.E.V.); (J.B.M.); (C.J.S.C.); (A.H.A.); (F.A.O.); (G.N.A.R.); (M.C.S.G.); (K.F.)
| | - Mariana P. Nucci
- LIM44—Hospital das Clínicas da Faculdade Medicina da Universidade de São Paulo, São Paulo 05403-000, SP, Brazil;
| | - Lionel F. Gamarra
- Hospital Israelita Albert Einstein, São Paulo 05652-000, SP, Brazil; (O.F.M.D.); (N.M.E.V.); (J.B.M.); (C.J.S.C.); (A.H.A.); (F.A.O.); (G.N.A.R.); (M.C.S.G.); (K.F.)
| |
Collapse
|
17
|
Liu Q, Song S, Liu L, Hong W. In Vivo Seeding of Amyloid-β Protein and Implications in Modeling Alzheimer's Disease Pathology. Biomolecules 2025; 15:571. [PMID: 40305318 PMCID: PMC12024744 DOI: 10.3390/biom15040571] [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: 02/26/2025] [Revised: 03/28/2025] [Accepted: 04/05/2025] [Indexed: 05/02/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by extracellular plaques containing amyloid β-protein (Aβ) and intracellular neurofibrillary tangles formed by tau. Cerebral Aβ accumulation initiates a noxious cascade that leads to irreversible neuronal degeneration and memory impairment in older adults. Recent advances in Aβ seeding studies offer a promising avenue for exploring the mechanisms underlying amyloid deposition and the complex pathological features of AD. However, the extent to which inoculated Aβ seeds can induce reproducible and reliable pathological manifestations remains unclear due to significant variability across studies. In this review, we will discuss several factors that contribute to the induction or acceleration of amyloid deposition and consequent pathologies. Specifically, we focus on the diversity of host animals, sources and recipe of Aβ seeds, and inoculating strategies. By integrating these key aspects, this review aims to offer a comprehensive perspective on Aβ seeding in AD and provide guidance for modeling AD pathogenesis through the exogenous introduction of Aβ seeds.
Collapse
Affiliation(s)
- Qianmin Liu
- School of Biomedical Sciences, Hunan University, Changsha 410082, China;
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
| | - Simin Song
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen 518055, China
| | - Lu Liu
- School of Biomedical Sciences, Hunan University, Changsha 410082, China;
| | - Wei Hong
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
| |
Collapse
|
18
|
Faraco G. Dietary salt, vascular dysfunction, and cognitive impairment. Cardiovasc Res 2025; 120:2349-2359. [PMID: 39429024 PMCID: PMC11976728 DOI: 10.1093/cvr/cvae229] [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: 05/29/2024] [Revised: 08/24/2024] [Accepted: 09/15/2024] [Indexed: 10/22/2024] Open
Abstract
Excessive salt consumption is a major health problem worldwide leading to serious cardiovascular events including hypertension, heart disease, and stroke. Additionally, high-salt diet has been increasingly associated with cognitive impairment in animal models and late-life dementia in humans. High-salt consumption is harmful for the cerebral vasculature, disrupts blood supply to the brain, and could contribute to Alzheimer's disease pathology. Although animal models have advanced our understanding of the cellular and molecular mechanisms, additional studies are needed to further elucidate the effects of salt on brain function. Furthermore, the association between excessive salt intake and cognitive impairment will have to be more thoroughly investigated in humans. Since the harmful effects of salt on the brain are independent by its effect on blood pressure, in this review, I will specifically discuss the evidence, available in experimental models and humans, on the effects of salt on vascular and cognitive function in the absence of changes in blood pressure. Given the strong effects of salt on the function of immune cells, I will also discuss the evidence linking salt consumption to gut immunity dysregulation with particular attention to the ability of salt to disrupt T helper 17 (Th17) cell homeostasis. Lastly, I will briefly discuss the data implicating IL-17A, the major cytokine produced by Th17 cells, in vascular dysfunction and cognitive impairment.
Collapse
Affiliation(s)
- Giuseppe Faraco
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| |
Collapse
|
19
|
Mishra R, Upadhyay A. An update on mammalian and non-mammalian animal models for biomarker development in neurodegenerative disorders. Cell Mol Life Sci 2025; 82:147. [PMID: 40192808 PMCID: PMC11977071 DOI: 10.1007/s00018-025-05668-y] [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: 11/16/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025]
Abstract
Neurodegeneration is one of the leading factor for death globally, affecting millions of people. Developing animal models are critical to understand biological processes and comprehend pathological hallmarks of neurodegenerative diseases. For decades, many animal models have served as excellent tools to determine the disease progression, develop diagnostic methods and design novel therapies against distinct pathologies. Here, we provide a comprehensive overview of both, mammalian and non-mammalian animal models, with a focus on three most common and aggressive neurodegenerative disorders: Alzheimer's disease, Parkinson's disease and Spinocerebellar ataxia-1. We highlight various approaches including transgene, gene transfer, and chemically-induced methods used to develop disease models. In particular, we discuss applications of both non-mammalian and mammalian contributions in research on neurodegeneration. It is exciting to learn the roles of animal models in disease pathomechanisms, identifying biomarkers and hence devising novel interventions to treat neuropathological conditions.
Collapse
Affiliation(s)
- Ribhav Mishra
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Arun Upadhyay
- Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Chhattisgarh, 491002, India
| |
Collapse
|
20
|
Antico O, Thompson PW, Hertz NT, Muqit MMK, Parton LE. Targeting mitophagy in neurodegenerative diseases. Nat Rev Drug Discov 2025; 24:276-299. [PMID: 39809929 DOI: 10.1038/s41573-024-01105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
Mitochondrial dysfunction is a hallmark of idiopathic neurodegenerative diseases, including Parkinson disease, amyotrophic lateral sclerosis, Alzheimer disease and Huntington disease. Familial forms of Parkinson disease and amyotrophic lateral sclerosis are often characterized by mutations in genes associated with mitophagy deficits. Therefore, enhancing the mitophagy pathway may represent a novel therapeutic approach to targeting an underlying pathogenic cause of neurodegenerative diseases, with the potential to deliver neuroprotection and disease modification, which is an important unmet need. Accumulating genetic, molecular and preclinical model-based evidence now supports targeting mitophagy in neurodegenerative diseases. Despite clinical development challenges, small-molecule-based approaches for selective mitophagy enhancement - namely, USP30 inhibitors and PINK1 activators - are entering phase I clinical trials for the first time.
Collapse
Affiliation(s)
- Odetta Antico
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Paul W Thompson
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK
| | | | - Miratul M K Muqit
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura E Parton
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK.
| |
Collapse
|
21
|
Sonsalla MM, Babygirija R, Johnson M, Cai S, Cole M, Yeh CY, Grunow I, Liu Y, Vertein D, Calubag MF, Trautman ME, Green CL, Rigby MJ, Puglielli L, Lamming DW. Acarbose ameliorates Western diet-induced metabolic and cognitive impairments in the 3xTg mouse model of Alzheimer's disease. GeroScience 2025; 47:1569-1591. [PMID: 39271570 PMCID: PMC11978593 DOI: 10.1007/s11357-024-01337-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Age is the greatest risk factor for Alzheimer's disease (AD) as well as for other disorders that increase the risk of AD such as diabetes and obesity. There is growing interest in determining if interventions that promote metabolic health can prevent or delay AD. Acarbose is an anti-diabetic drug that not only improves glucose homeostasis, but also extends the lifespan of wild-type mice. Here, we test the hypothesis that acarbose will not only preserve metabolic health, but also slow or prevent AD pathology and cognitive deficits in 3xTg mice, a model of AD, fed either a Control diet or a high-fat, high-sucrose Western diet (WD). We find that acarbose decreases the body weight and adiposity of WD-fed 3xTg mice, increasing energy expenditure while also stimulating food consumption, and improves glycemic control. Both male and female WD-fed 3xTg mice have worsened cognitive deficits than Control-fed mice, and these deficits are ameliorated by acarbose treatment. Molecular and histological analysis of tau and amyloid pathology identified sex-specific effects of acarbose which are uncoupled from the dramatic improvements in cognition in females, suggesting that the benefits of acarbose on AD may be largely driven by improved metabolic health. In conclusion, our results suggest that acarbose may be a promising intervention to prevent, delay, or even treat AD, especially in individuals consuming a WD.
Collapse
Affiliation(s)
- Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Madeline Johnson
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Samuel Cai
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mari Cole
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Yang Liu
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Diana Vertein
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michaela E Trautman
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael J Rigby
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Ave, MFCB Rm 4147, Madison, WI, 53705, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, 53705, USA.
- University of Wisconsin-Madison Comprehensive Diabetes Center, Madison, WI, 53705, USA.
| |
Collapse
|
22
|
Zhu G, Zhang H, Xie R, Younis MR, Fu S, Wang X, Liu B, Li K, Lui S, Wu M. Monitoring Acidification Preceding Aβ Deposition in Alzheimer's Disease. Adv Healthc Mater 2025; 14:e2404907. [PMID: 40103521 DOI: 10.1002/adhm.202404907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/08/2025] [Indexed: 03/20/2025]
Abstract
Amyloid beta (Aβ) is the primary early biomarker of Alzheimer's disease (AD), and since an acidic environment promotes Aβ aggregation, acidification plays a crucial role in AD progression. In this study, a novel acid-responsive near-infrared (NIR) fluorescent probe alongside multiple molecular biology techniques to investigate the temporal relationship between acidification and Aβ deposition, as well as the underlying mechanisms of acidification is employed. By monitoring 2- to 11-month-old APP/PS1 mice and wild-type (WT) mice, it is detected significant fluorescence signal in APP/PS1 mice beginning at 3 months preceding Aβ deposition at 5 months, and peaking at 5 months, followed by cognitive deficits at 8 months. Additionally, elevated monocarboxylate transporter 4 (MCT4) protein expression in 3-month-old APP/PS1 mice indicated disruption of astrocyte-neuron lactate shuttle (ANLS) homeostasis. Overall, this findings first demonstrate that acidification precedes Aβ deposition, peaks at the onset of Aβ deposition, and diminishes thereafter, with early acidification likely driven by the disruption of ANLS.
Collapse
Affiliation(s)
- Guannan Zhu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Hong Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Ruoxi Xie
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Muhammad Rizwan Younis
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California at Los Angeles, Los Angeles, California, 90095, USA
| | - Shengxiang Fu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Xiaoze Wang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Beibei Liu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, No. 29, Wangjiang Road, Chengdu, 610064, China
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| |
Collapse
|
23
|
Yeapuri P, Machhi J, Foster EG, Kadry R, Bhattarai S, Lu Y, Sil S, Sapkota R, Srivastava S, Kumar M, Ikezu T, Poluektova LY, Gendelman HE, Mosley RL. Amyloid precursor protein and presenilin-1 knock-in immunodeficient mice exhibit intraneuronal Aβ pathology, microgliosis, and extensive neuronal loss. Alzheimers Dement 2025; 21:e70084. [PMID: 40195277 PMCID: PMC11975631 DOI: 10.1002/alz.70084] [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: 11/15/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 04/09/2025]
Abstract
INTRODUCTION Transgenic mice overexpressing familial Alzheimer's disease (AD) mutations (FAD) show non-physiological traits, and their immunocompetent backgrounds limit their use in AD immunotherapy research. Preclinical models that reflect human immune responses in AD are needed. METHODS Using CRISPR-Cas9, we developed single (NA) and double (NAPS) knock-in (KI) amyloid precursor protein (APP)KM670,671NL (Swedish) and presenilin 1 (PS 1)M146VFAD mutations on an immunodeficient NOG (NOD.Cg-PrkdcscidIl2rgtm1Sug/JicTac) background. The models were confirmed by Sanger sequencing and evaluated for AD-like pathology. RESULTS Both NA and NAPS mice developed pathology without overexpression artifacts. Mutation-induced upregulation of APP-CTF-β led to intraneuronal human amyloid beta (Aβ) (6E10) deposits and amyloid-associated microgliosis as early as 3 months, which increased with age. The addition of the PS 1M146V mutation doubled the amyloid load. The models displayed broad neuronal loss, resulting in brain atrophy in older mice. DISCUSSION These models replicate intraneuronal amyloid pathology and, with human immune reconstitution potential, enable novel studies of human immune responses in AD. HIGHLIGHTS A novel Alzheimer's disease (AD) knock-in (KI) mouse was developed and characterized on an immunodeficient NOG background. The model provides a platform for human immune studies and the evaluation of immunotherapies for AD. The KI mice demonstrate intraneuronal Aβ deposits and amyloid-associated microglial reactions. KI mice demonstrate extensive neuronal loss. Human immune reconstitution enables studies of infectious AD co-morbidities, such as the human immunodeficiency and herpes simplex viruses.
Collapse
Affiliation(s)
- Pravin Yeapuri
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Jatin Machhi
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Emma G. Foster
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Rana Kadry
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Yaman Lu
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Susmita Sil
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Roshan Sapkota
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Shefali Srivastava
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Mohit Kumar
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Tsuneya Ikezu
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
| | - Larisa Y. Poluektova
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Rodney Lee Mosley
- Department of Pharmacology and Experimental NeuroscienceCenter for Neurodegenerative DisordersCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
24
|
Liu S, Liu X, Ke M, Wang J. Sleep fragmentation impairs cognitive function and exacerbates Alzheimer's disease-related pathology in a mouse model by disrupting mitochondrial biogenesis. Exp Neurol 2025; 386:115153. [PMID: 39832661 DOI: 10.1016/j.expneurol.2025.115153] [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: 09/25/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
A large proportion of Alzheimer's disease (AD) patients suffer from various types of chronic sleep disturbances, including sleep fragmentation (SF). In addition, impaired mitochondrial biogenesis is an important feature of AD, but whether it is altered in sleep disorders has not been fully elucidated. Hence, we aimed to investigate the relationship between SF and mitochondrial biogenesis and the possible impact of SF on AD-related pathology. In this study, thirty-six 9-month-old 3xTgAD model mice and thirty-six 9-month-old wild-type (WT) C57BL/6 J mice were divided into a control group (6 weeks of normal sleep), a SF group (6 weeks of SF) and a SF + recovery sleep group (6 weeks of SF followed by 2 weeks of recovery sleep). Cognitive functions were assessed by behavioural experiments. Mitochondrial structure and function and the activity of a classic mitochondrial biogenesis signalling pathway were investigated using transmission electron microscopy (TEM), reverse transcription quantitative polymerase chain reaction (RT-qPCR), immunofluorescence and Western blotting. Markers of AD-related pathology, including the levels of amyloid β (Aβ) and tau proteins, were assessed by immunofluorescence and Western blotting. The expression of insulin-degrading enzyme (IDE) was assessed by Western blotting. We found that long-term SF impaired the cognitive functions of the mice. In addition, chronic SF reduced the expression of mitochondrial respiratory chain components, the number of mitochondria, the fluorescence intensity of COX-IV, the level of mitochondrial DNA (mtDNA) and the expression of crucial regulators of the AMPK/SIRT-1/PGC-1α signalling pathway in the mouse prefrontal cortex and hippocampus, while recovery sleep could partly abrogate these effects. Moreover, SF reduced the protein level of IDE and increased the Aβ burden and tau hyperphosphorylation. This study demonstrates that chronic SF can negatively regulate the AMPK/SIRT-1/PGC-1α signalling pathway to disrupt mitochondrial biogenesis in the brains of mice, which may subsequently exacerbate AD-related pathology by decreasing the expression of IDE.
Collapse
Affiliation(s)
- Shunjie Liu
- Department of Neurology, The Second People's Hospital of Foshan, Foshan 528000, Guangdong Province, China
| | - Xingyi Liu
- Department of Neurology, The Second People's Hospital of Foshan, Foshan 528000, Guangdong Province, China
| | - Man Ke
- Department of Neurology, The Second People's Hospital of Foshan, Foshan 528000, Guangdong Province, China
| | - Jinliang Wang
- Department of Neurology, The Second People's Hospital of Foshan, Foshan 528000, Guangdong Province, China.
| |
Collapse
|
25
|
Bansal VA, Tan JM, Soon HR, Zainolabidin N, Saido T, Ch'ng TH. Aβ-driven nuclear pore complex dysfunction alters activation of necroptosis proteins in a mouse model of Alzheimer's disease. eLife 2025; 13:RP92069. [PMID: 40132021 PMCID: PMC11936419 DOI: 10.7554/elife.92069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
The emergence of Aβ pathology is one of the hallmarks of Alzheimer's disease (AD), but the mechanisms and impact of Aβ in progression of the disease is unclear. The nuclear pore complex (NPC) is a multi-protein assembly in mammalian cells that regulates movement of macromolecules across the nuclear envelope; its function is shown to undergo age-dependent decline during normal aging and is also impaired in multiple neurodegenerative disorders. Yet not much is known about the impact of Aβ on NPC function in neurons. Here, we examined NPC and nucleoporin (NUP) distribution and nucleocytoplasmic transport using a mouse model of AD (AppNL-G-F/NL-G-F) that expresses Aβ in young animals. Our studies revealed that a time-dependent accumulation of intracellular Aβ corresponded with a reduction of NPCs and NUPs in the nuclear envelope which resulted in the degradation of the permeability barrier and inefficient segregation of nucleocytoplasmic proteins, and active transport. As a result of the NPC dysfunction App KI neurons become more vulnerable to inflammation-induced necroptosis - a programmed cell death pathway where the core components are activated via phosphorylation through nucleocytoplasmic shutting. Collectively, our data implicates Aβ in progressive impairment of nuclear pore function and further confirms that the protein complex is vulnerable to disruption in various neurodegenerative diseases and is a potential therapeutic target.
Collapse
Affiliation(s)
| | - Jia Min Tan
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- School of Biological Science, Nanyang Technological UniversitySingaporeSingapore
| | - Hui Rong Soon
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- School of Biological Science, Nanyang Technological UniversitySingaporeSingapore
| | | | - Takaomi Saido
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- School of Biological Science, Nanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
26
|
Hu B, Shi Y, Xiong F, Chen YT, Zhu X, Carrillo E, Wen X, Drolet N, Rajpurohit C, Xu X, Lee DF, Soto C, Zhong S, Jayaraman V, Zheng H, Li W. Rewired m6A methylation of promoter antisense RNAs in Alzheimer's disease regulates global gene transcription in the 3D nucleome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.22.644756. [PMID: 40196645 PMCID: PMC11974732 DOI: 10.1101/2025.03.22.644756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
N6-methyladenosine (m6A) is the most prevalent internal RNA modification that can impact mRNA expression post-transcriptionally. Recent progress indicates that m6A also acts on nuclear or chromatin-associated RNAs to impact transcriptional and epigenetic processes. However, the landscapes and functional roles of m6A in human brains and neurodegenerative diseases, including Alzheimer's disease (AD), have been under-explored. Here, we examined RNA m6A methylome using total RNA-seq and meRIP-seq in middle frontal cortex tissues of post-mortem human brains from individuals with AD and age-matched counterparts. Our results revealed AD-associated alteration of m6A methylation on both mRNAs and various noncoding RNAs. Notably, a series of promoter antisense RNAs (paRNAs) displayed cell-type-specific expression and changes in AD, including one produced adjacent to the MAPT locus that encodes the Tau protein. We found that MAPT-paRNA is enriched in neurons, and m6A positively controls its expression. In iPSC-derived human excitatory neurons, MAPT-paRNA promotes expression of hundreds of genes related to neuronal and synaptic functions, including a key AD resilience gene MEF2C, and plays a neuroprotective role against excitotoxicity. By examining RNA-DNA interactome in the three-dimensional (3D) nuclei of human brains, we demonstrated that brain paRNAs can interact with both cis- and trans-chromosomal target genes to impact their transcription. These data together reveal previously unexplored landscapes and functions of noncoding RNAs and m6A methylome in brain gene regulation, neuronal survival and AD pathogenesis.
Collapse
Affiliation(s)
- Benxia Hu
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Yuqiang Shi
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Feng Xiong
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Yi-Ting Chen
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center and UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Xiaoyu Zhu
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Elisa Carrillo
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Xingzhao Wen
- Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Nathan Drolet
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Chetan Rajpurohit
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, USA
| | - Dung-Fang Lee
- The University of Texas MD Anderson Cancer Center and UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Claudio Soto
- The University of Texas MD Anderson Cancer Center and UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Sheng Zhong
- Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Vasanthi Jayaraman
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center and UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Wenbo Li
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center and UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
27
|
Sun R, Han M, Lin Y, Ma S, Tu H, Yang X, Zhang F, Zhang HT. Inhibition of PDE4B ameliorates cognitive defects in the model of alcoholic dementia in 3xTg-AD mice via PDE4B/cAMP/PKA signaling. Int J Neuropsychopharmacol 2025; 28:pyaf009. [PMID: 39921664 PMCID: PMC11923544 DOI: 10.1093/ijnp/pyaf009] [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: 07/24/2024] [Accepted: 02/07/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Chronic, heavy alcohol use may lead to permanent brain damage, cognitive impairment, and dementia. One of the most serious consequences is alcoholic dementia (AlD). Phosphodiesterase-4 (PDE4) inhibitors have been shown to exhibit beneficial effects on cognition deficits and alcoholism. However, it is not known whether PDE4 inhibitors can be used to treat AlD. A33, a relatively selective PDE4B inhibitor, is absent of the emetic effect associated with PDE4D. The effect of A33 on memory and cognition in AlD remains unclear. METHODS We investigated the effects of A33 and the PDE4 inhibitor rolipram on memory and cognition using an AlD animal model, that is, APP/PS1/Tau mice drinking alcohol in the 2-bottle choice test, with or without A33 or rolipram treatment for 3 weeks. The animal groups were compared in behavioral tests related to learning and memory. Neurochemical measures were conducted to explore the underlying mechanism of A33. RESULTS Compared to wild-type controls, AlD mice showed impairments of learning ability and memory in the behavior tests; this was attenuated by treatment of rolipram or A33. In addition, administration of rolipram or A33 in AlD mice further alleviated neuropathological alterations in the hippocampus, including Aβ expression and deposition; rolipram or A33 also decreased the levels of inflammatory cytokines, including interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), as well as nuclear factor kappa-B (NF-κB). Further, rolipram or A33 decreased the activation of microglia while increased cyclic adenosine monophosphate (cAMP) levels in the hippocampus of AlD mice. CONCLUSIONS These results revealed that the alleviation of the cognitive impairment of AlD in APP/PS1/Tau triple transgenic mice by rolipram or A33 was linked to the action of the PDE4B/cAMP/PKA signaling pathway. A33 can be a promising therapeutic agent for AlD-related cognitive dysfunction.
Collapse
Affiliation(s)
- Rongzhen Sun
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Mei Han
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Yuanyuan Lin
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Shengyao Ma
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Huan Tu
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Xueliang Yang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Fang Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Han-Ting Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| |
Collapse
|
28
|
Palacino F, Manganotti P, Benussi A. Targeting Neural Oscillations for Cognitive Enhancement in Alzheimer's Disease. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:547. [PMID: 40142358 PMCID: PMC11943909 DOI: 10.3390/medicina61030547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025]
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia, is marked by progressive cognitive decline, affecting memory, language, orientation, and behavior. Pathological hallmarks include extracellular amyloid plaques and intracellular tau tangles, which disrupt synaptic function and connectivity. Neural oscillations, the rhythmic synchronization of neuronal activity across frequency bands, are integral to cognitive processes but become dysregulated in AD, contributing to network dysfunction and memory impairments. Targeting these oscillations has emerged as a promising therapeutic strategy. Preclinical studies have demonstrated that specific frequency modulations can restore oscillatory balance, improve synaptic plasticity, and reduce amyloid and tau pathology. In animal models, interventions, such as gamma entrainment using sensory stimulation and transcranial alternating current stimulation (tACS), have shown efficacy in enhancing memory function and modulating neuroinflammatory responses. Clinical trials have reported promising cognitive improvements with repetitive transcranial magnetic stimulation (rTMS) and deep brain stimulation (DBS), particularly when targeting key hubs in memory-related networks, such as the default mode network (DMN) and frontal-parietal network. Moreover, gamma-tACS has been linked to increased cholinergic activity and enhanced network connectivity, which are correlated with improved cognitive outcomes in AD patients. Despite these advancements, challenges remain in optimizing stimulation parameters, individualizing treatment protocols, and understanding long-term effects. Emerging approaches, including transcranial pulse stimulation (TPS) and closed-loop adaptive neuromodulation, hold promise for refining therapeutic strategies. Integrating neuromodulation with pharmacological and lifestyle interventions may maximize cognitive benefits. Continued interdisciplinary efforts are essential to refine these approaches and translate them into clinical practice, advancing the potential for neural oscillation-based therapies in AD.
Collapse
Affiliation(s)
| | | | - Alberto Benussi
- Neurology Unit, Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (F.P.); (P.M.)
| |
Collapse
|
29
|
Sánchez-Garrido Campos G, Zafra ÁM, Estévez-Rodríguez M, Cordones I, Ruffini G, Márquez-Ruiz J. Preclinical insights into gamma-tACS: foundations for clinical translation in neurodegenerative diseases. Front Neurosci 2025; 19:1549230. [PMID: 40143845 PMCID: PMC11936909 DOI: 10.3389/fnins.2025.1549230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Gamma transcranial alternating current stimulation (gamma-tACS) represents a novel neuromodulation technique with promising therapeutic applications across neurodegenerative diseases. This mini-review consolidates recent preclinical and clinical findings, examining the mechanisms by which gamma-tACS influences neural oscillations, enhances synaptic plasticity, and modulates neuroimmune responses. Preclinical studies have demonstrated the capacity of gamma-tACS to synchronize neuronal firing, support long-term neuroplasticity, and reduce markers of neuroinflammation, suggesting its potential to counteract neurodegenerative processes. Early clinical studies indicate that gamma-tACS may improve cognitive functions and network connectivity, underscoring its ability to restore disrupted oscillatory patterns central to cognitive performance. Given the intricate and multifactorial nature of gamma oscillations, the development of tailored, optimized tACS protocols informed by extensive animal research is crucial. Overall, gamma-tACS presents a promising avenue for advancing treatments that support cognitive resilience in a range of neurodegenerative conditions.
Collapse
Affiliation(s)
| | - Ángela M. Zafra
- Department of Physiology, Anatomy and Cell Biology, Pablo de Olavide University, Seville, Spain
| | - Marta Estévez-Rodríguez
- Department of Physiology, Anatomy and Cell Biology, Pablo de Olavide University, Seville, Spain
| | - Isabel Cordones
- Department of Physiology, Anatomy and Cell Biology, Pablo de Olavide University, Seville, Spain
| | - Giulio Ruffini
- Brain Modeling Department, Neuroelectrics Barcelona, Barcelona, Spain
| | - Javier Márquez-Ruiz
- Department of Physiology, Anatomy and Cell Biology, Pablo de Olavide University, Seville, Spain
| |
Collapse
|
30
|
Yao J, Li K, Fu Z, Zheng J, Chen Z, Xu J, Lai G, Huang Y, Huang J, You G, Han S, He Z, Liu Q, Li N. Human tau promotes Warburg effect-like glycolytic metabolism under acute hyperglycemia conditions. J Biol Chem 2025; 301:108376. [PMID: 40054691 PMCID: PMC12018107 DOI: 10.1016/j.jbc.2025.108376] [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: 01/30/2025] [Accepted: 02/26/2025] [Indexed: 04/13/2025] Open
Abstract
The neurofilaments formed by hyperphosphorylated tau is a hallmark of tauopathies. However, the biological functions of tau and the physiological significance of its phosphorylation are still not fully understood. By using human tau (441 a.a.) transgenic (hTau) mice, murine tau KO mice, and C57BL/6J (C57) mice, unexpectedly, we found that under acute hyperglycemia conditions, JNK but not previously reported GSK3β mediated tau phosphorylation. Moreover, Akt, the inhibitory kinase upstream of GSK3β, was activated in a tau-dependent manner. Furthermore, under acute high glucose conditions, the presence of human tau significantly augmented Akt activation but inhibited 4E-BP1 phosphorylation simultaneously, indicating that human tau is also involved in regulating the alternative activation of mTORC1/2. By comparing the hippocampal membrane-associated proteome, we found that human tau influenced the homeostasis of protein-membrane association under acute hyperglycemia conditions. Of note, with respect to C57 and Tau KO mice, the membrane association of oxidative phosphorylation-related proteins was impeded by human tau in the hippocampus. In vitro study consistently showed that aerobic glycolysis was promoted in the presence of human tau under high glucose conditions, which maintained the ratio of NAD+/NADH. On the other hand, human tau restricted the level of oxidative phosphorylation, modulated the activity of SDH, and reduced ROS production upon high glucose challenging. In summary, the current study revealed that human tau played an important role in regulating glycolytic metabolism under acute hyperglycemia conditions, which is similar with the Warburg effect, through influencing the homeostasis of protein-membrane association.
Collapse
Affiliation(s)
- Jinyi Yao
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Keying Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Zhenli Fu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Jingjing Zheng
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Zicong Chen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Jiahao Xu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Guoqing Lai
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Yaomin Huang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Jinsheng Huang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Guanying You
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Shuangxue Han
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Zhijun He
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Nan Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Brain Disease and Big Data Research Institute, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China; Instrumental Analysis Center of Shenzhen University, Shenzhen University, Shenzhen, China; Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
31
|
Yao M, Rosario ER, Soper JC, Pike CJ. Androgens Regulate Tau Phosphorylation Through Phosphatidylinositol 3-Kinase-Protein Kinase B-Glycogen Synthase Kinase 3β Signaling. Neuroscience 2025; 568:503-518. [PMID: 35777535 PMCID: PMC9797620 DOI: 10.1016/j.neuroscience.2022.06.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/11/2022] [Accepted: 06/23/2022] [Indexed: 12/31/2022]
Abstract
Age-related testosterone depletion in men is a risk factor for Alzheimer's disease (AD). How testosterone modulates AD risk remains to be fully elucidated, although regulation of tau phosphorylation has been suggested as a contributing protective action. To investigate the relationship between testosterone and tau phosphorylation, we first evaluated the effect of androgen status on tau phosphorylation in 3xTg-AD mice. Depletion of endogenous androgens via gonadectomy resulted in increased tau phosphorylation that was prevented by acute testosterone treatment. Parallel alterations in the phosphorylation of both glycogen synthase kinase 3β (GSK3β) and protein kinase B (Akt) suggest possible components of the underlying signaling pathway. To further explore mechanism, primary cultured neurons were treated with a physiological concentration of testosterone or its active metabolite dihydrotestosterone (DHT). Results showed that testosterone and DHT induced significant decreases in phosphorylated tau and significant increases in phosphorylation of Akt and GSK3β. Pharmacological inhibition of phosphatidylinositol 3-kinase (PI3K) effectively inhibited androgen-induced increases in Akt and GSK3β phosphorylation, and decreases in tau phosphorylation. In addition, androgen receptor (AR) knock-down by small interfering RNA prevented androgen-induced changes in the phosphorylation of Akt, GSK3β and tau, suggesting an AR-dependent mechanism. Additional experiments demonstrated androgen-induced changes in Akt, GSK3β and tau phosphorylation in AR-expressing PC12 cells but not in AR-negative PC12 cells. Together, these results suggest an AR-dependent pathway involving PI3K-Akt-GSK3β signaling through which androgens can reduce tau phosphorylation. These findings identify an additional protective mechanism of androgens that can improve neural health and inhibit development of AD.
Collapse
Affiliation(s)
- Mingzhong Yao
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Emily R Rosario
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Jenna Carroll Soper
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Christian J Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
32
|
Zhong M, Xu QQ, Huang MQ, Zhan RT, Huang XQ, Yang W, Lin ZX, Xian YF. Rhynchophylline alleviates cognitive deficits in multiple transgenic mouse models of Alzheimer's disease via modulating neuropathology and gut microbiota. Acta Pharmacol Sin 2025:10.1038/s41401-025-01475-0. [PMID: 40011632 DOI: 10.1038/s41401-025-01475-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/02/2025] [Indexed: 02/28/2025]
Abstract
Amyloid-beta (Aβ) aggregation, phosphorylated tau accumulation and neuroinflammation are considered as three hallmarks of Alzheimer's disease (AD). Rhynchophylline (RN), the major alkaloid of a Chinese medicinal plant Uncaria rhynchophylla, has been shown to possess potent anti-AD effects. This study explored the effects of RN on Aβ pathology, tauopathy, and neuroinflammation using three AD mouse models, including TgCRND8, 3×Tg-AD, and 5×FAD, with RN treatment lasting for 4, 6, and 6 months, respectively, followed by behavioral tests and biological assays. In addition, BV2 cells were employed to further evaluate the biological effects of RN. RN treatment improved cognitive functions by reducing anxiety-like behaviors, enhancing recognition ability, and ameliorating learning impairments. It modulated Aβ processing through reducing the Aβ-producing enzyme activities and enhancing degradation enzyme activities, thereby diminishing Aβ accumulation. RN also decreased hyperphosphorylated tau proteins at Thr181, Thr205, Ser396, and Ser404 sites. Moreover, RN diminished neuroinflammation by reducing microglia and astrocyte activation and lowering the release of inflammatory cytokines. Furthermore, RN treatment could restore gut microbiota dysbiosis in 5×FAD mice. In BV2 cells, knockdown of p53, HDAC2, and Galectin-3 markedly enhanced the anti-inflammatory effects of RN. Overall, the anti-AD properties of RN were attributed to its regulation of multiple biological pathways, including regulation of the p53/PINK1 signaling pathway, inhibition of the HDAC2/AMPK signaling pathway, suppression of the Galectin-3/C/EBPβ/AEP signaling pathway, and modulation of gut microflora dysbiosis. This pioneering study unambiguously revealed the effects of RN on cognitive impairments, APP processing, tauopathy, and neuroinflammation in different transgenic mouse models with differing AD burdens, highlighting its potential as an anti-AD therapeutic agent and enhancing the scientific basis for its clinical use in treating AD.
Collapse
Affiliation(s)
- Mei Zhong
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Qing-Qing Xu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Ming-Qing Huang
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Ruo-Ting Zhan
- Key Laboratory of Chinese Medicinal Resource from Lingnan (Ministry of Education), School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiao-Qi Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wen Yang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong, Shatin, N.T., Hong Kong SAR, China.
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| |
Collapse
|
33
|
Huang Y, Zhai Y, Zhao D, Wu M, Shen Q, Zhao W, Wang Q, Yao L, Li W. UHPLC-Q Exactive-Orbitrap-MS and network pharmacology analyses to investigate the mechanism by which Danggui-Shaoyao-San affects 27-OHC-induced cell damage in SH-SY5Y/C6 coculture. BMC Complement Med Ther 2025; 25:75. [PMID: 39994624 PMCID: PMC11849221 DOI: 10.1186/s12906-025-04751-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: 03/21/2024] [Accepted: 01/07/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Danggui-Shaoyao-San (DSS) is a classic Chinese medicine formula that has been extensively studied for its efficacy in treating Alzheimer's disease (AD). However, its mechanism of action is still unclear. METHODS In this study, UHPLC-Q Exactive-Orbitrap-MS was used to analyze and identify the compounds in DSS. Network pharmacology was used to analyze the common targets of drug-containing serum chemistries and AD, as well as the AD pathways in which drug-containing serum chemistries may be involved. The 27-OHC-induced SH-SY5Y/C6 coculture cell injury model was used to explore the mechanism of action of DSS in the treatment of AD. RESULTS UHPLC-Q Exactive-Orbitrap-MS analysis identified 73 chemical constituents in DSS aqueous extract and 39 compounds in drug-containing serum. According to network pharmacology analysis, DSS and AD share 181 common targets, with interleukin-6 (IL-6) and tumor necrosis factor (TNF) being the main effective targets. Furthermore, DSS may treat AD through the modulation of lipid metabolism-related pathways and the interleukin-17 (IL-17) signaling pathway. 27-hydroxycholesterol acid (27-OHC) significantly reduced the viability of SH-SY5Y cells and C6 cells in vitro, while DSS administration upregulated the expression of cytochrome P450 46A1 (CYP46A1) and cytochrome P450 7B1 (CYP7B1) enzymes and reduced cholesterol levels in SH-SY5Y cells. Additionally, DSS decreased reactive oxygen species (ROS) levels and increased glutathione (GSH) levels in coculture systems. DSS downregulated the expression of IL-17 in 27-OHC-injured SH-SY5Y cells and downregulated the expression of TNF-α, IL-6 and transforming growth factor-β1 (TGF-β1) in 27-OHC-injured C6 cells. CONCLUSION This study revealed the effective components, targets and mechanisms of DSS in the treatment of AD, highlighting the significant potential of DSS in treating this disease.
Collapse
Affiliation(s)
- Yi Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingying Zhai
- School of Pharmacy, Xinyang Agriculture and Forestry University, Henan, China
| | - Di Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mingan Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Shen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Limei Yao
- School of Traditional Chinese Medicine Healthcare, Guangdong Food and Drug Vocational College, Tianhe District, 321 Longdong North Road, Guangzhou, 510520, China.
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
34
|
Silva J, Tumurbaatar B, Guptarak J, Zhang WR, Fracassi A, Taglialatela G. Sub-Immunosuppressive Tacrolimus Ameliorates Amyloid-Beta and Tau Pathology in 3xTg-AD Mice. Int J Mol Sci 2025; 26:1797. [PMID: 40076425 PMCID: PMC11898583 DOI: 10.3390/ijms26051797] [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: 01/14/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Tacrolimus (TAC) has emerged as a potential therapy for Alzheimer's disease (AD), with the challenge of balancing its therapeutic benefits against its immunosuppressive effects. This study explores the efficacy of a sub-immunosuppressive TAC dosing regimen to ameliorate AD-related pathologies. TAC was administered daily for 14 days, with drug concentrations measured via liquid chromatography tandem mass spectrometry (LC-MS/MS) in whole blood and hippocampal tissue from C57BL6J mice, while immunofluorescence analyses and Western blotting (performed on hippocampal extracts) were conducted in 10-12 month old 3xTg-AD mice to evaluate levels of tau and amyloid-beta (Aβ) proteins. The results from LC-MS/MS revealed that lower TAC doses resulted in sub-immunosuppressive blood levels, while still penetrating the hippocampi. Immunofluorescence showed reductions in tau and Aβ proteins in 3xTg-AD mice. Additionally, Western blot analyses revealed reductions in tau and Aβ, along with increases in synaptic and autophagy-related proteins. These findings highlight the potential of sub-immunosuppressive TAC doses in effectively targeting AD pathology while minimizing the risk of chronic systemic immunosuppression. Further research and clinical trials are warranted to establish the optimal TAC dosing regimen for AD treatment.
Collapse
Affiliation(s)
- Jacqueline Silva
- Mitchell Center for Neurodegenerative Disease, Department of Neurology, The University of Texas Medical Branch at Galveston, Galveston, TX 77550, USA; (J.S.); (B.T.); (J.G.); (W.-R.Z.); (A.F.)
| | - Batbayar Tumurbaatar
- Mitchell Center for Neurodegenerative Disease, Department of Neurology, The University of Texas Medical Branch at Galveston, Galveston, TX 77550, USA; (J.S.); (B.T.); (J.G.); (W.-R.Z.); (A.F.)
| | - Jutatip Guptarak
- Mitchell Center for Neurodegenerative Disease, Department of Neurology, The University of Texas Medical Branch at Galveston, Galveston, TX 77550, USA; (J.S.); (B.T.); (J.G.); (W.-R.Z.); (A.F.)
| | - Wen-Ru Zhang
- Mitchell Center for Neurodegenerative Disease, Department of Neurology, The University of Texas Medical Branch at Galveston, Galveston, TX 77550, USA; (J.S.); (B.T.); (J.G.); (W.-R.Z.); (A.F.)
| | - Anna Fracassi
- Mitchell Center for Neurodegenerative Disease, Department of Neurology, The University of Texas Medical Branch at Galveston, Galveston, TX 77550, USA; (J.S.); (B.T.); (J.G.); (W.-R.Z.); (A.F.)
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Disease, Department of Neurology, The University of Texas Medical Branch at Galveston, Galveston, TX 77550, USA; (J.S.); (B.T.); (J.G.); (W.-R.Z.); (A.F.)
- The Moody Brain Health Institute, The University of Texas Medical Branch at Galveston, Galveston, TX 77550, USA
| |
Collapse
|
35
|
Han X, Li PH, Wang S, Blakely T, Aggarwal S, Gopalani B, Sanchez M, Schalek R, Meirovitch Y, Lin Z, Berger D, Wu Y, Aly F, Bay S, Delatour B, Lafaye P, Pfister H, Wei D, Jain V, Ploegh H, Lichtman J. Mapping Alzheimer's Molecular Pathologies in Large-Scale Connectomics Data: A Publicly Accessible Correlative Microscopy Resource. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.24.563674. [PMID: 37961104 PMCID: PMC10634883 DOI: 10.1101/2023.10.24.563674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Connectomics using volume-electron-microscopy enables mapping and analysis of neuronal networks, revealing insights into neural circuit function and dysfunction. In Alzheimer's disease (AD), where amyloid-β (Aβ) and hyperphosphorylated-Tau (pTau) are implicated, connectomics offers an approach to unravel how these molecules contribute to circuit alterations by enabling the study of these molecules within the context of the complete local neuronal and glial milieu. We present a volumetric-correlated-light-and-electron microscopy (vCLEM) protocol using fluorescent nanobodies to localize Aβ and pTau within a large-scale connectomics dataset from the hippocampus of the 3xTg AD mouse model. A key outcome of this work is a publicly accessible vCLEM dataset, featuring fluorescent labeling of Aβ and pTau in the ultrastructural context with segmented neurons, glia, and synapses. This dataset provides a unique resource for exploring AD pathology in the context of connectomics and fosters collaborative opportunities in neurodegenerative disease research. As a proof-of-principle, we uncovered new localizations of Aβ and pTau, including pTau-positive spine-like protrusions at the axon initial segment and changes in the number and size of synapses near Aβ plaques. Our vCLEM approach facilitates the discovery of both molecular and structural alterations within large-scale EM data, advancing connectomics research in Alzheimer's and other neurodegenerative diseases.
Collapse
|
36
|
Zhou F, Lian W, Yuan X, Wang Z, Xia C, Yan Y, Wang W, Tong Z, Cheng Y, Xu J, He J, Zhang W. Cornuside alleviates cognitive impairments induced by Aβ 1-42 through attenuating NLRP3-mediated neurotoxicity by promoting mitophagy. Alzheimers Res Ther 2025; 17:47. [PMID: 39972387 PMCID: PMC11837312 DOI: 10.1186/s13195-025-01695-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder in which mitochondrial dysfunction and neuroinflammation play crucial roles in its progression. Our previous studies found that cornuside from Cornus officinalis Sieb.Et Zucc is an anti-AD candidate, however, its underlying mechanism remains unknown. In the present study, AD mice were established by intracerebroventricular injection of Aβ1-42 and treated with cornuside (3, 10, 30 mg/kg) for 2 weeks. Cornuside significantly ameliorated behavioral deficits, protected synaptic plasticity and relieved neuronal damage in Aβ1-42 induced mice. Importantly, cornuside decreased NLRP3 inflammasome activation, characterized by decreased levels of NLRP3, ASC, Caspase-1, GSDMD, and IL-1β. Furthermore, cornuside promoted mitophagy accompanied by decreasing SQSTM1/p62 and promoting LC3B-I transforming into LC3B-II, via Pink1/Parkin signaling instead of FUNDC1 or BNIP3 pathways. In order to investigate the relationship between NLRP3 inflammasome and mitophagy in the neuroprotective mechanism of cornuside, we established an in-vitro model in BV2 cells exposed to LPS and Aβ1-42. And cornuside inhibited NLRP3 inflammasome activation and subsequent cytokine release, also protected neurons from damaging factors in microenvironment of conditional culture. Cornuside improved mitochondrial function by promoting oxidative phosphorylation and glycolysis, decreasing the production of ROS and mitochondrial membrane potential depolarization. Besides, mitophagy was also facilitated with increased colocalization of MitoTracker with LC3B and Parkin, and Pink1/Parkin, FUNDC1 and BNIP3 pathways were all involved in the mechanism of cornuside. By blocking the formation of autophagosomes by 3-MA, the protective effects on mitochondria, the inhibition on NLRP3 inflammasome as well as neuronal protection in conditional culture were eliminated. There is reason to believe that the promotion of mitophagy plays a key role in the NLRP3 inhibition of cornuside. In conclusion, cornuside re-establishes the mitophagy flux which eliminates damaged mitochondria and recovers mitochondrial function, both of them are in favor of inhibiting NLRP3 inflammasome activation, then alleviating neuronal and synaptic damage, and finally improving cognitive function.
Collapse
Affiliation(s)
- Fulin Zhou
- Institute of Clinical Medical Sciences, Department of Pharmacy, China-Japan, Friendship Hospital 2nd, Yinghua Dongjie, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Wenwen Lian
- Institute of Clinical Medical Sciences, Department of Pharmacy, China-Japan, Friendship Hospital 2nd, Yinghua Dongjie, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Xiaotang Yuan
- School of Life Science, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Zexing Wang
- School of Life Science, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Congyuan Xia
- Institute of Clinical Medical Sciences, Department of Pharmacy, China-Japan, Friendship Hospital 2nd, Yinghua Dongjie, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Yu Yan
- Institute of Clinical Medical Sciences, Department of Pharmacy, China-Japan, Friendship Hospital 2nd, Yinghua Dongjie, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Wenping Wang
- Institute of Clinical Medical Sciences, Department of Pharmacy, China-Japan, Friendship Hospital 2nd, Yinghua Dongjie, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Zhuohang Tong
- School of Life Science, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Yungchi Cheng
- Department of Pharmacology, School of Medicine, Yale University, Connecticut, New Haven, USA
| | - Jiekun Xu
- School of Life Science, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, People's Republic of China.
| | - Jun He
- Institute of Clinical Medical Sciences, Department of Pharmacy, China-Japan, Friendship Hospital 2nd, Yinghua Dongjie, Chaoyang District, Beijing, 100029, People's Republic of China.
| | - Weiku Zhang
- Institute of Clinical Medical Sciences, Department of Pharmacy, China-Japan, Friendship Hospital 2nd, Yinghua Dongjie, Chaoyang District, Beijing, 100029, People's Republic of China.
| |
Collapse
|
37
|
Vuic B, Milos T, Kvak E, Konjevod M, Tudor L, Farkas S, Nedic Erjavec G, Nikolac Perkovic M, Zelena D, Svob Strac D. Neuroprotective Effects of Dehydroepiandrosterone Sulphate Against Aβ Toxicity and Accumulation in Cellular and Animal Model of Alzheimer's Disease. Biomedicines 2025; 13:432. [PMID: 40002846 PMCID: PMC11853520 DOI: 10.3390/biomedicines13020432] [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: 01/15/2025] [Revised: 02/04/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Beneficial effects of neurosteroid dehydroepiandrosterone sulphate (DHEAS) on cognition, emotions and behavior have been previously reported, suggesting its potential in the prevention and treatment of various neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease (AD). This study aimed to investigate the potential neuroprotective actions of DHEAS against Aβ toxicity in both cellular and animal models of AD. Methods: After optimizing the AD model in vitro, we investigated the DHEAS effects on the viability and death of primary mouse neurons exposed to toxic Aβ42 oligomers for 24 h. In order to extend our research to an in vivo study, we further tested the acute effects of intraperitoneal DHEAS administration on the Aβ plaque density in different brain regions of 3xTg-AD mice, an animal model of AD. Results: In cell culture, DHEAS hampered the decrease in the neuronal viability caused by toxic Aβ oligomers, primarily by influencing mitochondrial function and apoptosis. DHEAS also counteracted the increase in the mRNA expression of selected genes (PI3K, Akt, Bcl2, Bax), induced in neuronal culture by treatment with Aβ42 oligomers. Obtained data suggested the involvement of mitochondria, caspases 3 and 7, as well as the PI3K/Akt and Bcl2 signaling network in the antiapoptotic properties of DHEAS in neurons. Forty-eight hours after DHEAS treatment, a significantly lower number of Aβ plaques was observed in the motor cortex but not in other brain areas of 3xTg-AD mice. Conclusions: Results indicated potential neuroprotective effects of DHEAS against Aβ toxicity and accumulation, suggesting that DHEAS supplementation should be further studied as a novel option for AD prevention and/or treatment.
Collapse
Affiliation(s)
- Barbara Vuic
- Laboratory for Molecular Neuropsychiatry, Ruder Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10 000 Zagreb, Croatia; (B.V.); (T.M.); (M.K.); (L.T.); (G.N.E.); (M.N.P.)
| | - Tina Milos
- Laboratory for Molecular Neuropsychiatry, Ruder Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10 000 Zagreb, Croatia; (B.V.); (T.M.); (M.K.); (L.T.); (G.N.E.); (M.N.P.)
| | - Erika Kvak
- Laboratory of Behavioural and Stress Studies, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (E.K.); (S.F.); (D.Z.)
| | - Marcela Konjevod
- Laboratory for Molecular Neuropsychiatry, Ruder Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10 000 Zagreb, Croatia; (B.V.); (T.M.); (M.K.); (L.T.); (G.N.E.); (M.N.P.)
| | - Lucija Tudor
- Laboratory for Molecular Neuropsychiatry, Ruder Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10 000 Zagreb, Croatia; (B.V.); (T.M.); (M.K.); (L.T.); (G.N.E.); (M.N.P.)
| | - Szidónia Farkas
- Laboratory of Behavioural and Stress Studies, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (E.K.); (S.F.); (D.Z.)
| | - Gordana Nedic Erjavec
- Laboratory for Molecular Neuropsychiatry, Ruder Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10 000 Zagreb, Croatia; (B.V.); (T.M.); (M.K.); (L.T.); (G.N.E.); (M.N.P.)
| | - Matea Nikolac Perkovic
- Laboratory for Molecular Neuropsychiatry, Ruder Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10 000 Zagreb, Croatia; (B.V.); (T.M.); (M.K.); (L.T.); (G.N.E.); (M.N.P.)
| | - Dora Zelena
- Laboratory of Behavioural and Stress Studies, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (E.K.); (S.F.); (D.Z.)
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Ruder Boskovic Institute, Division of Molecular Medicine, Bijenicka cesta 54, 10 000 Zagreb, Croatia; (B.V.); (T.M.); (M.K.); (L.T.); (G.N.E.); (M.N.P.)
| |
Collapse
|
38
|
Mengr A, Šmotková Z, Pačesová A, Železná B, Kuneš J, Maletínská L. Reduction of Neuroinflammation as a Common Mechanism of Action of Anorexigenic and Orexigenic Peptide Analogues in the Triple Transgenic Mouse Model of Alzheimer´s Disease. J Neuroimmune Pharmacol 2025; 20:18. [PMID: 39932627 PMCID: PMC11813825 DOI: 10.1007/s11481-025-10174-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/24/2025] [Indexed: 02/14/2025]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Characterized by progressive neurodegeneration, AD typically begins with mild cognitive decline escalating to severe impairment in communication and responsiveness. It primarily affects cerebral regions responsible for cognition, memory, and language processing, significantly impeding the functional independence of patients. With nearly 50 million dementia cases worldwide, a number expected to triple by 2050, the need for effective treatments is more urgent than ever. Recent insights into the association between obesity, type 2 diabetes mellitus, and neurodegenerative disorders have led to the development of promising treatments involving antidiabetic and anti-obesity agents. One such novel promising candidate for addressing AD pathology is a lipidized analogue of anorexigenic peptide called prolactin-releasing peptide (palm11-PrRP31). Interestingly, anorexigenic and orexigenic peptides have opposite effects on food intake regulation, however, both types exhibit neuroprotective properties. Recent studies have also identified ghrelin, an orexigenic peptide, as a potential neuroprotective agent. Hence, we employed both anorexigenic and orexigenic compounds to investigate the common mechanisms underpinning their neuroprotective effects in a triple transgenic mouse model of AD (3xTg-AD mouse model) combining amyloid-beta (Aβ) pathology and Tau pathology, two hallmarks of AD. We treated 3xTg-AD mice for 4 months with two stable lipidized anorexigenic peptide analogues - palm11-PrRP31, and liraglutide, a glucagon-like peptide 1 (GLP-1) analogue - as well as Dpr3-ghrelin, a stable analogue of the orexigenic peptide ghrelin, and using the method of immunohistochemistry and western blot demonstrate the effects of these compounds on the development of AD-like pathology in the brain. Palm11-PrRP31, Dpr3-ghrelin, and liraglutide reduced intraneuronal deposits of Aβ plaque load in the hippocampi and amygdalae of 3xTg-AD mice. Palm11-PrRP31 and Dpr3-ghrelin reduced microgliosis in the hippocampi, amygdalae, and cortices of 3xTg-AD mice. Palm11-PrRP31 and liraglutide reduced astrocytosis in the amygdalae of 3xTg-AD mice. We propose that these peptides are involved in reducing inflammation, a common mechanism underlying their therapeutic effects. This is the first study to demonstrate improvements in AD pathology following the administration of both orexigenic and anorexigenic compounds, highlighting the therapeutic potential of food intake-regulating peptides in neurodegenerative disorders.
Collapse
Affiliation(s)
- Anna Mengr
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| | - Zuzana Šmotková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Kateřinská 32, 12108, Prague, Czech Republic
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 00, Prague, Czech Republic
| | - Andrea Pačesová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic
- Institute of Physiology of the Czech Academy of Sciences, Vídeňská 1083, 142 00, Prague, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nám. 2, 160 00, Prague, Czech Republic.
| |
Collapse
|
39
|
Yarbro JM, Han X, Dasgupta A, Yang K, Liu D, Shrestha HK, Zaman M, Wang Z, Yu K, Lee DG, Vanderwall D, Niu M, Sun H, Xie B, Chen PC, Jiao Y, Zhang X, Wu Z, Chepyala SR, Fu Y, Li Y, Yuan ZF, Wang X, Poudel S, Vagnerova B, He Q, Tang A, Ronaldson PT, Chang R, Yu G, Liu Y, Peng J. Human and mouse proteomics reveals the shared pathways in Alzheimer's disease and delayed protein turnover in the amyloidome. Nat Commun 2025; 16:1533. [PMID: 39934151 PMCID: PMC11814087 DOI: 10.1038/s41467-025-56853-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
Murine models of Alzheimer's disease (AD) are crucial for elucidating disease mechanisms but have limitations in fully representing AD molecular complexities. Here we present the comprehensive, age-dependent brain proteome and phosphoproteome across multiple mouse models of amyloidosis. We identified shared pathways by integrating with human metadata and prioritized components by multi-omics analysis. Collectively, two commonly used models (5xFAD and APP-KI) replicate 30% of the human protein alterations; additional genetic incorporation of tau and splicing pathologies increases this similarity to 42%. We dissected the proteome-transcriptome inconsistency in AD and 5xFAD mouse brains, revealing that inconsistent proteins are enriched within amyloid plaque microenvironment (amyloidome). Our analysis of the 5xFAD proteome turnover demonstrates that amyloid formation delays the degradation of amyloidome components, including Aβ-binding proteins and autophagy/lysosomal proteins. Our proteomic strategy defines shared AD pathways, identifies potential targets, and underscores that protein turnover contributes to proteome-transcriptome discrepancies during AD progression.
Collapse
Affiliation(s)
- Jay M Yarbro
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xian Han
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Abhijit Dasgupta
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Computer Science and Engineering, SRM University AP, Andhra Pradesh, India
| | - Ka Yang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Danting Liu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Him K Shrestha
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Masihuz Zaman
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhen Wang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kaiwen Yu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Dong Geun Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - David Vanderwall
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mingming Niu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Huan Sun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Boer Xie
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ping-Chung Chen
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yun Jiao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xue Zhang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhiping Wu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Surendhar R Chepyala
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yingxue Fu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yuxin Li
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zuo-Fei Yuan
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xusheng Wang
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Suresh Poudel
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Barbora Vagnerova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Qianying He
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Andrew Tang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Rui Chang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Gang Yu
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University School of Medicine, West Haven, CT, USA
- Department of Biomedical Informatics & Data Science, Yale University School of Medicine, West Haven, CT, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
40
|
Cantone AF, Burgaletto C, Di Benedetto G, Gaudio G, Giallongo C, Caltabiano R, Broggi G, Bellanca CM, Cantarella G, Bernardini R. Rebalancing Immune Interactions within the Brain-Spleen Axis Mitigates Neuroinflammation in an Aging Mouse Model of Alzheimer's Disease. J Neuroimmune Pharmacol 2025; 20:15. [PMID: 39918606 PMCID: PMC11805801 DOI: 10.1007/s11481-025-10177-7] [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: 10/15/2024] [Accepted: 01/24/2025] [Indexed: 02/11/2025]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide, characterized by accumulation of amyloid-β protein and hyperphosphorylated tau protein in the brain. Neuroinflammation, resulting from chronic activation of brain-resident innate immune cells as well as enhanced peripheral leukocyte access across the blood-brain barrier, crucially affects AD progression. In this context, TNFSF10, a cytokine substantially expressed in the AD brain, has been shown to modulate both the innate and the adaptive branches of the immune response in AD-related neuroinflammation. In this study, we explored whether a TNFSF10-neutralizing treatment could represent a tool to re-balance the overall overshooting inflammatory response in a mouse model of AD. Specifically, 3xTg-AD mice were treated sub-chronically with an anti-TNFSF10 monoclonal antibody for three months, and were then sacrificed at 15 months. TNFSF10 neutralization reduced the expression of the inflammatory marker CD86, inversely related to levels of the anti-inflammatory marker CD206 in the brain of 3xTg-AD mice, suggesting a switch of microglia towards a neuroprotective phenotype. Similar results were observed in the splenic macrophage population. Moreover, flow cytometry revealed a significant decrease of CD4+CD25+FOXP3+ T regulatory cells as well as reduced number of CD11b+LY6Chigh proinflammatory monocytes in both the brain and the spleen of 3xTg-AD mice treated with anti-TNFSF10 monoclonal antibody. Finally, the treatment resulted in lower count of splenic CD4+ and CD8+ T cells expressing PD1. The data suggest that TNFSF10 system-targeted treatment effectively restrain overshooting central and peripheral inflammation by rebalancing the overall immune response, mitigating the progression of AD pathology.
Collapse
Affiliation(s)
- Anna Flavia Cantone
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Chiara Burgaletto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Giulia Di Benedetto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
- Clinical Toxicology Unit, University Hospital of Catania, Catania, Italy.
| | - Gabriella Gaudio
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Cesarina Giallongo
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", Division of Hematology, University of Catania, Catania, Italy
| | - Rosario Caltabiano
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", Anatomic Pathology, University of Catania, Catania, Italy
| | - Giuseppe Broggi
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", Anatomic Pathology, University of Catania, Catania, Italy
| | - Carlo Maria Bellanca
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
- Clinical Toxicology Unit, University Hospital of Catania, Catania, Italy
| | - Giuseppina Cantarella
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
- Clinical Toxicology Unit, University Hospital of Catania, Catania, Italy
| |
Collapse
|
41
|
Martínez-Orozco H, Bencomo-Martínez A, Maya-Arteaga JP, Rubio-De Anda PF, Sanabria-Romero F, Casas ZGM, Rodríguez-Vargas I, Hernández-Puga AG, Sablón-Carrazana M, Menéndez-Soto del Valle R, Rodríguez-Tanty C, Díaz-Cintra S. CNEURO-201, an Anti-amyloidogenic Agent and σ1-Receptor Agonist, Improves Cognition in the 3xTg Mouse Model of Alzheimer's Disease by Multiple Actions in the Pathology. Int J Mol Sci 2025; 26:1301. [PMID: 39941068 PMCID: PMC11818425 DOI: 10.3390/ijms26031301] [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: 12/16/2024] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
The complexity of Alzheimer's disease (AD) pathophysiology represents a significant challenge in the development of effective therapeutic agents for its treatment. CNEURO-201 (CN, also Amylovis-201) is a novel pharmaceutical agent with dual activity as an anti-amyloid-β (Aβ) agent and σ1 receptor agonist. CN exhibits great efficacy at very low doses, delaying cognitive impairment and alleviating Aβ load in animal models of AD. However, CN functions on other remains related to this pathology remain to be investigated. The present study sought to evaluate the effects of CN treatment at a dosage of 0.1 mg kg-1 (p.o) over an eight-week period in the 3xTg-AD mouse model. In silico studies, as well as biochemical and immunofluorescence assays, were conducted on brain tissue to investigate the CN effects on acetylcholine metabolism, redox system, and glial cell activation-related biomarkers in brain regions that are relevant for memory. The results demonstrated that CN effectively rescues cognitive impairment of 3xTg-AD mice by influencing glial activity to reduce existing Aβ plaques but also modulating acetylcholine metabolism and the enzymatic response of proteins involved in the redox system. Our outcomes reinforced the potential of CN in treating AD by acting on multiple pathways altered in this disease.
Collapse
Affiliation(s)
- Humberto Martínez-Orozco
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Alberto Bencomo-Martínez
- Departamento de Farmacología, Centro de Neurociencias de Cuba, Avenida Independencia 8126, La Habana 11600, Cuba; (A.B.-M.); (M.S.-C.); (R.M.-S.d.V.); (C.R.-T.)
| | - Juan Pablo Maya-Arteaga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Pedro Francisco Rubio-De Anda
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Fausto Sanabria-Romero
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Zyanya Gloria Mena Casas
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Isaac Rodríguez-Vargas
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| | - Ana Gabriela Hernández-Puga
- Centro de Investigación Biomédica Avanzada, Facultad de Medicina, Universidad Autónoma de Querétaro, Carretera a Chichimequillas S/N, Santiago de Querétaro 76140, Querétaro, Mexico;
| | - Marquiza Sablón-Carrazana
- Departamento de Farmacología, Centro de Neurociencias de Cuba, Avenida Independencia 8126, La Habana 11600, Cuba; (A.B.-M.); (M.S.-C.); (R.M.-S.d.V.); (C.R.-T.)
| | - Roberto Menéndez-Soto del Valle
- Departamento de Farmacología, Centro de Neurociencias de Cuba, Avenida Independencia 8126, La Habana 11600, Cuba; (A.B.-M.); (M.S.-C.); (R.M.-S.d.V.); (C.R.-T.)
| | - Chryslaine Rodríguez-Tanty
- Departamento de Farmacología, Centro de Neurociencias de Cuba, Avenida Independencia 8126, La Habana 11600, Cuba; (A.B.-M.); (M.S.-C.); (R.M.-S.d.V.); (C.R.-T.)
| | - Sofía Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología-UNAM Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla 76230, Querétaro, Mexico; (H.M.-O.); (J.P.M.-A.); (P.F.R.-D.A.); (F.S.-R.); (Z.G.M.C.); (I.R.-V.)
| |
Collapse
|
42
|
Watamura N, Foiani MS, Bez S, Bourdenx M, Santambrogio A, Frodsham C, Camporesi E, Brinkmalm G, Zetterberg H, Patel S, Kamano N, Takahashi M, Rueda-Carrasco J, Katsouri L, Fowler S, Turkes E, Hashimoto S, Sasaguri H, Saito T, Islam AS, Benner S, Endo T, Kobayashi K, Ishida C, Vendruscolo M, Yamada M, Duff KE, Saido TC. In vivo hyperphosphorylation of tau is associated with synaptic loss and behavioral abnormalities in the absence of tau seeds. Nat Neurosci 2025; 28:293-307. [PMID: 39719507 PMCID: PMC11802456 DOI: 10.1038/s41593-024-01829-7] [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: 05/28/2023] [Accepted: 10/23/2024] [Indexed: 12/26/2024]
Abstract
Tau pathology is a hallmark of several neurodegenerative diseases, including frontotemporal dementia and Alzheimer's disease. However, the sequence of events and the form of tau that confers toxicity are still unclear, due in large part to the lack of physiological models of tauopathy initiation and progression in which to test hypotheses. We have developed a series of targeted mice expressing frontotemporal-dementia-causing mutations in the humanized MAPT gene to investigate the earliest stages of tauopathy. MAPTInt10+3G>A and MAPTS305N;Int10+3G>A lines show abundant hyperphosphorylated tau in the hippocampus and entorhinal cortex, but they do not develop seed-competent fibrillar structures. Accumulation of hyperphosphorylated tau was accompanied by neurite degeneration, loss of viable synapses and indicators of behavioral abnormalities. Our results demonstrate that neuronal toxicity can occur in the absence of fibrillar, higher-order structures and that tau hyperphosphorylation is probably involved in the earliest etiological events in tauopathies showing isoform ratio imbalance.
Collapse
Affiliation(s)
- Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan.
- UK Dementia Research Institute at University College London, London, UK.
| | - Martha S Foiani
- UK Dementia Research Institute at University College London, London, UK.
| | - Sumi Bez
- UK Dementia Research Institute at University College London, London, UK
| | - Mathieu Bourdenx
- UK Dementia Research Institute at University College London, London, UK
| | - Alessia Santambrogio
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Claire Frodsham
- UK Dementia Research Institute at University College London, London, UK
| | - Elena Camporesi
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology,The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology,The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, London, UK
- Queen Square Institute of Neurology, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology,The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Saisha Patel
- UK Dementia Research Institute at University College London, London, UK
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Mika Takahashi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | | | - Loukia Katsouri
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | - Stephanie Fowler
- UK Dementia Research Institute at University College London, London, UK
- Nuffield Department of Medicine, Oxford-GSK Institute of Molecular and Computational Medicine, Centre for Human Genetics, Oxford, UK
| | - Emir Turkes
- UK Dementia Research Institute at University College London, London, UK
| | - Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Pioneering Research Division, Medical Innovation Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
- Dementia Pathophysiology Collaboration Unit, RIKEN Center for Brain Science, Wako, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Afm Saiful Islam
- Queen Square Institute of Neurology, University College London, London, UK
| | - Seico Benner
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
| | | | - Katsuji Kobayashi
- Department of Psychiatry, Awazu Neuropsychiatric Hospital, Ishikawa, Japan
| | - Chiho Ishida
- Department of Neurology, NHO Iou National Hospital, Iwade-machi, Japan
| | - Michele Vendruscolo
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, UK
| | - Masahito Yamada
- Department of Internal Medicine, Division of Neurology, Kudanzaka Hospital, Tokyo, Japan
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo, Japan
- Kanazawa University, Kanazawa, Japan
| | - Karen E Duff
- UK Dementia Research Institute at University College London, London, UK.
- Queen Square Institute of Neurology, University College London, London, UK.
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan.
| |
Collapse
|
43
|
Calvin-Dunn KN, Mcneela A, Leisgang Osse A, Bhasin G, Ridenour M, Kinney JW, Hyman JM. Electrophysiological insights into Alzheimer's disease: A review of human and animal studies. Neurosci Biobehav Rev 2025; 169:105987. [PMID: 39732222 DOI: 10.1016/j.neubiorev.2024.105987] [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: 01/22/2024] [Revised: 11/16/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
This review highlights the crucial role of neuroelectrophysiology in illuminating the mechanisms underlying Alzheimer's disease (AD) pathogenesis and progression, emphasizing its potential to inform the development of effective treatments. Electrophysiological techniques provide unparalleled precision in exploring the intricate networks affected by AD, offering insights into the synaptic dysfunction, network alterations, and oscillatory abnormalities that characterize the disease. We discuss a range of electrophysiological methods, from non-invasive clinical techniques like electroencephalography and magnetoencephalography to invasive recordings in animal models. By drawing on findings from these studies, we demonstrate how electrophysiological research has deepened our understanding of AD-related network disruptions, paving the way for targeted therapeutic interventions. Moreover, we underscore the potential of electrophysiological modalities to play a pivotal role in evaluating treatment efficacy. Integrating electrophysiological data with clinical neuroimaging and longitudinal studies holds promise for a more comprehensive understanding of AD, enabling early detection and the development of personalized treatment strategies. This expanded research landscape offers new avenues for unraveling the complexities of AD and advancing therapeutic approaches.
Collapse
Affiliation(s)
- Kirsten N Calvin-Dunn
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Cleveland Clinic Lou Ruvo Center for Brain Health, United States.
| | - Adam Mcneela
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States
| | - A Leisgang Osse
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Brain Health, University of Nevada, Las Vegas, United States
| | - G Bhasin
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Psychology, University of Nevada, Las Vegas, United States
| | - M Ridenour
- Department of Psychology, University of Nevada, Las Vegas, United States
| | - J W Kinney
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Brain Health, University of Nevada, Las Vegas, United States
| | - J M Hyman
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Psychology, University of Nevada, Las Vegas, United States
| |
Collapse
|
44
|
Moreno-Rodríguez M, Martínez-Gardeazabal J, Bengoetxea de Tena I, Llorente-Ovejero A, Lombardero L, González de San Román E, Giménez-Llort L, Manuel I, Rodríguez-Puertas R. Cognitive improvement via cortical cannabinoid receptors and choline-containing lipids. Br J Pharmacol 2025; 182:1038-1058. [PMID: 39489624 DOI: 10.1111/bph.17381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND AND PURPOSE Recent research linking choline-containing lipids to degeneration of basal forebrain cholinergic neurons in neuropathological states illustrates the challenge of balancing lipid integrity with optimal acetylcholine levels, essential for memory preservation. The endocannabinoid system influences learning and memory processes regulated by cholinergic neurotransmission. Therefore, we hypothesised that activation of the endocannabinoid system may confer neuroprotection against cholinergic degeneration. EXPERIMENTAL APPROACH We examined the neuroprotective potential of sub-chronic treatments with the cannabinoid agonist WIN55,212-2, using ex vivo organotypic tissue cultures including nucleus basalis magnocellularis and cortex and in vivo rat models of specific cholinergic damage induced by 192IgG-saporin. Levels of lipids, choline and acetylcholine were measured with histochemical and immunofluorescence assays, along with [35S]GTPγS autoradiography of cannabinoid and muscarinic GPCRs and MALDI-mass spectrometry imaging analysis. Learning and memory were assessed by the Barnes maze and the novel object recognition test in rats and in the 3xTg-AD mouse model. KEY RESULTS Degeneration, induced by 192IgG-saporin, of baso-cortical cholinergic pathways resulted in memory deficits and decreased cortical levels of lysophosphatidylcholines (LPC). WIN55,212-2 restored cortical cholinergic transmission and LPC levels via activation of cannabinoid receptors. This activation altered cortical lipid homeostasis mainly by reducing sphingomyelins in lesioned animals. These modifications were crucial for memory recovery. CONCLUSION AND IMPLICATIONS We hypothesise that WIN55,212-2 facilitates an alternative choline source by breaking down sphingomyelins, leading to elevated cortical acetylcholine levels and LPCs. These results imply that altering choline-containing lipids via activation of cannabinoid receptors presents a promising therapeutic approach for dementia linked to cholinergic dysfunction.
Collapse
Affiliation(s)
- Marta Moreno-Rodríguez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jonatan Martínez-Gardeazabal
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Iker Bengoetxea de Tena
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Alberto Llorente-Ovejero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Laura Lombardero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | - Lydia Giménez-Llort
- Department of Psychiatry and Forensic Medicine, School of Medicine & Institute of Neuroscience, Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Iván Manuel
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Rafael Rodríguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Barakaldo, Spain
| |
Collapse
|
45
|
Jáuregui GV, Parpura V. Neuron-Astrocyte Interactions in Aging and Alzheimer's Disease: Dysregulation of Amyloid Precursor Protein. AGEING & LONGEVITY 2025; 6:117-128. [PMID: 40098995 PMCID: PMC11911455 DOI: 10.47855/jal9020-2025-2-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Amyloid precursor protein (APP) is central to Alzheimer's disease (AD) by its role in Aβ build-up and in neuronal and astrocytic malfunction. The major risk factor for late-onset AD is aging, which increases APP processing in both neurons and astrocytes, and consequently increases Aβ production. This focused review covers the subjects of how aging and AD affect APP dynamics within the both cell types and how astrocytes dysfunction can enhance neuroinflammation and neuronal dysfunction and injury. We discuss the interplay between neurons and astrocytes in aging and AD brains, where bi-directional cellular interactions accelerate neurodegeneration.
Collapse
Affiliation(s)
- Gretsen Velezmoro Jáuregui
- International Translational Neuroscience Research Institute, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Vladimir Parpura
- International Translational Neuroscience Research Institute, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
46
|
Eo H, Kim SH, Ju IG, Lee JH, Oh MS, Kim YJ. NXP032 Improves Memory Impairment Through Suppression of Tauopathy in PS19 Mice and Attenuates Okadaic Acid-Induced Tauopathy in SH-SY5Y Cells. J Neuroimmune Pharmacol 2025; 20:10. [PMID: 39891801 DOI: 10.1007/s11481-025-10175-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Tauopathy is widely observed in multiple neurodegenerative diseases such as Alzheimer's disease (AD) and characterized by abnormal tau protein phosphorylation, aggregation and its accumulation as a form of neurofibrillary tangle (NFT) in the brain. However, there are no effective treatments targeting tau pathology in the AD. Vitamin C is known to reduce tauopathy and modulate one of its regulators called glycogen synthase kinase 3 (GSK3) in the body. Nevertheless, vitamin C has a limitation of its stability during metabolism due to its chemical properties. Thus, in the current study, NXP032 (a vitamin C/aptamer complex) was tested as a candidate for tau-targeting treatment because it can preserve antioxidative efficacy of vitamin C before it can reach the target tissue. In this context, the current study aimed to investigate the therapeutic effect of NXP032 on tauopathy in vivo and in vitro. As a result, NXP032 attenuated cognitive and memory decline and reduced NFT and tau hyperphosphorylation in the P301S mutant human tau transgenic mice (or called PS19 mice). In addition, NXP032 suppressed neuroinflammation found in the PS19 mice. Furthermore, NXP032 protected SH-SY5Y human neuroblastoma cells from okadaic acid (OKA)-induced cytotoxicity. Especially, 10 ng/ml of NXP032 reduced tau hyperphosphorylation and GSK3 activation though its phosphorylation at Tyr216 site which were promoted by OKA treatment in the SH-SY5Y cells. Taken together, our results suggest that NXP032 might be a potential therapy for AD and tauopathy-related neurodegenerative disorders as well.
Collapse
Affiliation(s)
- Hyeyoon Eo
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Food and Nutrition, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Seong Hye Kim
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - In Gyoung Ju
- Department of Oriental Pharmaceutical Science, College of Pharmacy and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Joo Hee Lee
- College of Nursing Science, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Myung Sook Oh
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Oriental Pharmaceutical Science, College of Pharmacy and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Youn-Jung Kim
- College of Nursing Science, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
47
|
Saternos HC, Forero KV, Meqdad MA, Buqaileh R, Sunderman CL, Gallagher G, Messer WS, Mohieldin AM, Mucci CA, Kumariya S, Osman IA, Burkett JP, AbouAlaiwi WA. Muscarinic acetylcholine receptor 3 localized to primary endothelial cilia regulates blood pressure and cognition. Sci Rep 2025; 15:3745. [PMID: 39885271 PMCID: PMC11782538 DOI: 10.1038/s41598-025-87212-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
We previously demonstrated that the inability of primary endothelial cilia to sense fluid shear stress can lead to nitric oxide (NO) deficiency and cause hypertension (HTN). Decreased biosynthesis of NO contributes to cerebral amyloid angiopathy in Alzheimer's disease (AD) patients through increased deposition of amyloid beta (Aβ). However, the molecular mechanisms underlying the pathogenesis of HTN and AD are incompletely understood. The objective of this study was to examine the pathophysiological roles of vascular primary cilia and muscarinic acetylcholine receptor 3 (CHRM3) in HTN and AD. We discovered, for the first time, that CHRM3 was localized to primary cilia of endothelial and cerebrovascular cells, and that CHRM3 expression was downregulated in cilialess cells. Moreover, CHRM3 activation enhanced cilia length and sensory function in terms of eNOS activation. To further examine the role of vascular CHRM3 in vivo, we showed that endothelial CHRM3 knockout was associated with increased BP and attenuated acetylcholine-mediated vascular relaxation. In addition, endothelial CHRM3 knockout resulted in altered fear behavior. This demonstrates the physiological significance of endothelial CHRM3 signaling and primary cilia-derived NO production as an important mechanism in the control of BP and cognition.
Collapse
Affiliation(s)
- Hannah C Saternos
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kathleen V Forero
- Department of Pharmacology and Experimental Therapeutics; MS 1015, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Health Education Building; Room 282E, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | - Mahmood A Meqdad
- Department of Pharmacology and Experimental Therapeutics; MS 1015, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Health Education Building; Room 282E, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | - Raghad Buqaileh
- Department of Pharmacology and Experimental Therapeutics; MS 1015, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Health Education Building; Room 282E, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | - Clare L Sunderman
- Department of Pharmacology and Experimental Therapeutics; MS 1015, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Health Education Building; Room 282E, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | - Gillian Gallagher
- Department of Pharmacology and Experimental Therapeutics; MS 1015, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Health Education Building; Room 282E, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | - William S Messer
- Department of Pharmacology and Experimental Therapeutics; MS 1015, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Health Education Building; Room 282E, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | - Ashraf M Mohieldin
- Master of Pharmaceutical Sciences Department, College of Graduate Studies, California Northstate University, Elk Grove, CA, USA
| | - Claudio A Mucci
- Department of Biological Sciences, College of Natural Sciences and Mathematics, The University of Toledo, Toledo, OH, USA
| | - Sanjana Kumariya
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, USA
| | - Islam A Osman
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, USA
| | - James P Burkett
- Department of Neurosciences, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, USA
| | - Wissam A AbouAlaiwi
- Department of Pharmacology and Experimental Therapeutics; MS 1015, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Health Education Building; Room 282E, 3000 Arlington Ave, Toledo, OH, 43614, USA.
| |
Collapse
|
48
|
Radhakrishnan K, Zhang Y, Mustapha O, Weigel TK, Upchurch CM, Tian X, Herbert F, Huang W, Leitinger N, Eyo UB, Ai H, Ferris HA. 7-ketocholesterol contributes to microglia-driven increases in astrocyte reactive oxygen species in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.19.633810. [PMID: 39868327 PMCID: PMC11761689 DOI: 10.1101/2025.01.19.633810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Oxidative stress is a prominent feature of Alzheimer's disease. Within this context, cholesterol undergoes oxidation, producing the pro-inflammatory product 7-ketocholesterol (7-KC). In this study, we observe elevated levels of 7-KC in the brains of the 3xTg mouse model of AD. To further understand the contribution of 7-KC on the oxidative environment, we developed a method to express a genetically encoded fluorescent hydrogen peroxide (H2O2) sensor in astrocytes, the primary source of cholesterol in the brain. With this sensor, we discovered that 7-KC increases H2O2 levels in astrocytes in vivo, but not when directly applied to astrocytes in vitro. Interestingly, when 7-KC was applied to a microglia cell line alone or mixed astrocyte and microglia cultures, it resulted in microglia activation and increased oxidative stress in astrocytes. Depletion of microglia from 3xTg mice resulted in reduced 7-KC in the brains of these mice. Taken together, these findings suggest that 7-KC, acting through microglia, contributes to increased astrocyte oxidative stress in AD. This study sheds light on the complex interplay between cholesterol oxidation, microglia activation, and astrocyte oxidative stress in the pathogenesis of AD.
Collapse
Affiliation(s)
- Kayalvizhi Radhakrishnan
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - Yiyu Zhang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Oluwaseun Mustapha
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Thaddeus K. Weigel
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Clint M. Upchurch
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Xiaodong Tian
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Franklin Herbert
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Wenyuan Huang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Ukpong B. Eyo
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Huiwang Ai
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Heather A. Ferris
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
49
|
Udo MSB, Zaccarelli-Magalhães J, Clemons GA, Citadin CT, Langman J, Smith DJ, Matuguma LH, Tesic V, Lin HW. Blockade of A 2AR improved brain perfusion and cognitive function in a mouse model of Alzheimer's disease. GeroScience 2025:10.1007/s11357-025-01526-8. [PMID: 39843732 DOI: 10.1007/s11357-025-01526-8] [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: 11/20/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that affects more than 6.2 million Americans aged 65 and older, particularly women. Along with AD's main hallmarks (formation of β-amyloid plaques and tau neurofibrillary tangles), there are vascular alterations that occurs in AD pathology. Adenosine A2 receptor (A2AR) is one of the key factors of brain vascular autoregulation and is overexpressed in AD patients. Our previous findings suggest that protein arginine methyltransferase 4 (PRMT4) is overexpressed in AD, which leads to decrease in cerebral blood flow in aged female 3xTg mice. We aimed to investigate the mechanism behind A2AR signaling in the regulation of brain perfusion and blood-brain barrier integrity in age and sex-dependent 3xTg mice, and if it is related to PRMT4. Istradefylline, a highly selective A2AR antagonist, was used to modulate A2AR signaling. Aged female 3xTg and C57BL/6 J mice were evaluated for brain perfusion (via laser speckle) and cognitive function (via open field, T-maze and novel object recognition). Our results suggest that modulation of A2AR signaling in aged female 3xTg increased cerebral perfusion by decreasing PRMT4 expression, restored the levels of APP and tau, maintained blood-brain barrier integrity by maintaining the expression of tight junction proteins, and preserved functional learning/memory.
Collapse
Affiliation(s)
- Mariana Sayuri Berto Udo
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Julia Zaccarelli-Magalhães
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Garrett Alan Clemons
- Department of Biomedical Science, West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | - Cristiane Teresinha Citadin
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Julia Langman
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Drew James Smith
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Luiz Henrique Matuguma
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA
| | - Vesna Tesic
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Hung Wen Lin
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, 6431 Fannin St., Houston, TX, 77030, USA.
| |
Collapse
|
50
|
Sha X, Lin J, Wu K, Lu J, Yu Z. The TRPV1-PKM2-SREBP1 axis maintains microglial lipid homeostasis in Alzheimer's disease. Cell Death Dis 2025; 16:14. [PMID: 39809738 PMCID: PMC11732990 DOI: 10.1038/s41419-024-07328-8] [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: 07/13/2024] [Revised: 12/08/2024] [Accepted: 12/27/2024] [Indexed: 01/30/2025]
Abstract
Microglia are progressively activated by inflammation and exhibit phagocytic dysfunction in the pathogenesis of neurodegenerative diseases. Lipid-droplet-accumulating microglia were identified in the aging mouse and human brain; however, little is known about the formation and role of lipid droplets in microglial neuroinflammation of Alzheimer's disease (AD). Here, we report a striking buildup of lipid droplets accumulation in microglia in the 3xTg mouse brain. Moreover, we observed significant upregulation of PKM2 and sterol regulatory element binding protein 1 (SREBP1) levels, which were predominantly localized in microglia of 3xTg mice. PKM2 dimerization was necessary for SREBP1 activation and lipogenesis of lipid droplet-accumulating microglia. RNA sequencing analysis of microglia isolated from 3xTg mice exhibited transcriptomic changes in lipid metabolism, innate inflammation, and phagocytosis dysfunction; these changes were improved with capsaicin-mediated pharmacological activation of TRPV1 via inhibition of PKM2 dimerization and reduction of SREBP1 activation. Lipid droplet-accumulating microglia exhibited increased mitochondrial injury accompanied by impaired mitophagy, which was abrogated upon of TRPV1 activation. Capsaicin also rescued neuronal loss, tau pathology, and memory impairment in 3xTg mice. Our study suggests that TRPV1-PKM2-SREBP1 axis regulation of microglia lipid metabolism could be a therapeutic approach to alleviate the consequences of AD.
Collapse
Affiliation(s)
- Xudong Sha
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiayuan Lin
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Kexin Wu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jia Lu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhihua Yu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|