1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Xie J, Van Hoecke L, Van Wonterghem E, Van Imschoot G, Andries V, Vereecke L, Vandenbroucke RE. The gut-brain axis in Alzheimer's disease is shaped by commensal gut microbiota derived extracellular vesicles. Gut Microbes 2025; 17:2501193. [PMID: 40355382 PMCID: PMC12077474 DOI: 10.1080/19490976.2025.2501193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/31/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025] Open
Abstract
Emerging clinical and experimental evidence highlight the involvement of gut microbiota in the onset and progression of neurodegenerative diseases such as Alzheimer's disease (AD) via neuroinflammatory processes along the gut-brain axis. Despite this, the precise mechanisms governing gut microbial involvement in AD remain elusive. In this study, we observed that AppNL-G-F AD mice raised under germ-free (GF) conditions, display a reduced amyloid-β (Aβ) pathology, accompanied by a shift in microglial cells toward a less inflammatory state and increased phagocytotic efficiency. In addition, we demonstrate that gut microbiota depletion can protect against synaptic deficits in AD mice. Notably, administering bacterial extracellular vesicles (bEVs), i.e. nano-sized particles packed with bacterial components, derived from fecal slurry from specific pathogen-free housed AppNL-G-F AD mice, reversed the effects of GF conditions on both microglial activation and Aβ plaque accumulation. These findings reveal for the first time that commensal gut microbiota-derived bEVs have a major impact on AD pathology progression.
Collapse
Affiliation(s)
- Junhua Xie
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Griet Van Imschoot
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Vanessa Andries
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent University, Ghent, Belgium
| | - Lars Vereecke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent University, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent University, Ghent, Belgium
| |
Collapse
|
3
|
Miyazaki H, Ide K, Yamaguchi H, Ogawa M, Aoki N, Katsuse O, Odawara T, Asami T. Peripheral lymphocyte counts and regional cerebral blood flow on brain SPECT correlate in Alzheimer's disease: A retrospective cross-sectional study. Brain Res 2025; 1859:149657. [PMID: 40280533 DOI: 10.1016/j.brainres.2025.149657] [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: 11/18/2024] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Given emerging evidence of immune involvement in Alzheimer's disease (AD), we aimed to clarify whether peripheral lymphocyte counts are associated with regional cerebral blood flow (rCBF) as measured by brain single-photon emission computed tomography (SPECT). Identifying such a relationship may help to establish early, accessible biomarkers of disease progression. METHODS Participants were 111 patients diagnosed with AD at the Yokohama City University Hospital Medical Center for Dementia Diseases between January 2021 and December 2023 and who underwent blood tests and brain SPECT. Sex, age, cognitive function tests, peripheral blood values, and rCBF on brain SPECT were investigated retrospectively. Neutrophil and lymphocyte counts were extracted from blood values. In addition, the rCBF in each region (frontal, parietal, temporal, occipital, and limbic system) was calculated from brain SPECT, and correlation analysis between lymphocytes and rCBF was performed. RESULTS Significant positive correlations between lymphocyte counts in peripheral blood and rCBF were found in all regions except the left frontal lobe. In particular, the correlation coefficient between rCBF and lymphocyte count was highest in the right temporal lobe (ρ = 0.311, P = 0.001). CONCLUSIONS Peripheral blood lymphocyte counts are positively related to rCBF on brain SPECT, and lymphocytes can be an early biomarker that can be tested inexpensively and easily. Limitations include the retrospective cross-sectional design and single-center setting, which preclude analysis of causality and changes over time.
Collapse
Affiliation(s)
- Hidehito Miyazaki
- Department of Psychiatry, Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; Yokohama Hoyu Hospital, 644-1 Kanegaya, Asahi-ku, Yokohama, Kanagawa 241-0812, Japan
| | - Keiko Ide
- Department of Psychiatry, Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; Department of Psychiatry, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan.
| | - Hiroyuki Yamaguchi
- Department of Psychiatry, Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; Department of Psychiatry, Yokohama Comprehensive Care Continuum, 1735 Toriyama-cho, Kohoku-ku, Yokohama, Kanagawa 222-0035, Japan
| | - Matsuyoshi Ogawa
- Radiation Department, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Naoya Aoki
- Department of Psychiatry, Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; Yokohama Hoyu Hospital, 644-1 Kanegaya, Asahi-ku, Yokohama, Kanagawa 241-0812, Japan
| | - Omi Katsuse
- Department of Psychiatry, Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; Yokohama Hoyu Hospital, 644-1 Kanegaya, Asahi-ku, Yokohama, Kanagawa 241-0812, Japan
| | - Toshinari Odawara
- Department of Psychiatry, Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan; Health Management Center, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Takeshi Asami
- Department of Psychiatry, Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| |
Collapse
|
4
|
Kshirsagar S, Alvir RV, Pradeepkiran JA, Reddy AP, Reddy PH. Therapeutic potential of DDQ in enhancing mitochondrial health and cognitive function in Late-Onset Alzheimer's disease. Mitochondrion 2025; 83:102036. [PMID: 40158867 DOI: 10.1016/j.mito.2025.102036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/15/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline, mitochondrial dysfunction, and neuroinflammation. This study evaluates the therapeutic potential of DDQ, a small molecule in the humanized Abeta knockin (hAbKI) mice that represents late-onset AD. Our findings demonstrate that DDQ treatment significantly improves cognitive performance as assessed through behavioral tests, including the rotarod, open field, Y-maze, and Morris water maze, compared to untreated hAbKI mice. At the molecular level, DDQ promoted mitochondrial biogenesis, as evidenced by enhanced expression of key proteins like PGC1α, NRF1, and TFAM. Additionally, DDQ treatment facilitated mitophagy, as indicated by elevated levels of PINK1 and Parkin, and reduced neuroinflammation, reflected by decreased Iba1 and GFAP levels. Transmission electron microscopy analysis revealed a marked improvement in mitochondrial morphology, with increased mitochondrial length and reduced mitochondrial numbers in DDQ-treated mice. Furthermore, DDQ treatment led to an increase in mitophagic vacuoles, suggesting that it effectively removes dysfunctional mitochondria. Taken together, for the first time, our study results support the potential of DDQ as a promising neuroprotective agent for late-onset AD, addressing mitochondrial dysfunction, neuroinflammation, and cognitive decline. Our study focused on developing small molecules that modulate mitophagy, mitochondrial dynamics and neuroinflammatory pathways for aging, AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Rainier Vladlen Alvir
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | - Arubala P Reddy
- Department of Nutrition, Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
5
|
Li Y, Zhao Q, Wang Y, Du W, Yang R, Wu J, Li Y. Lipid droplet accumulation in microglia and their potential roles. Lipids Health Dis 2025; 24:215. [PMID: 40514678 PMCID: PMC12166618 DOI: 10.1186/s12944-025-02633-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS), where lipid metabolism is critical for maintaining homeostasis. In response to various external stimuli, they demonstrate a range of phenotypic expressions and lipid metabolic reprogramming. Lipid droplets (LDs) are dynamic organelles that function beyond energy storage, actively participating in neuropathological progress. Recent investigations have identified a subset of microglia characterized by the accumulation of LDs, referred to as "lipid-droplet-accumulating microglia" (LDAM). This review aims to investigate the processes involved in LD formation and degradation, the factors that modulate them, focusing particularly on the function of LDAM and their implications for CNS disorders. By synthesizing current evidence, we clarify the biological significance of LDs in these cells and their therapeutic targeting potential, providing new directions for future research.
Collapse
Affiliation(s)
- Yunxia Li
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Qi Zhao
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Yan Wang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
- School of Health Medicine, Nantong Institute of Technology, Nantong, Jiangsu, China
| | - Wenyi Du
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Riyun Yang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Jian Wu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China.
| | - Yi Li
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
6
|
Prakash P, Manchanda P, Paouri E, Bisht K, Sharma K, Rajpoot J, Wendt V, Hossain A, Wijewardhane PR, Randolph CE, Chen Y, Stanko S, Gasmi N, Gjojdeshi A, Card S, Fine J, Jethava KP, Clark MG, Dong B, Ma S, Crockett A, Thayer EA, Nicolas M, Davis R, Hardikar D, Allende D, Prayson RA, Zhang C, Davalos D, Chopra G. Amyloid-β induces lipid droplet-mediated microglial dysfunction via the enzyme DGAT2 in Alzheimer's disease. Immunity 2025; 58:1536-1552.e8. [PMID: 40393454 PMCID: PMC12168635 DOI: 10.1016/j.immuni.2025.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/30/2024] [Accepted: 04/22/2025] [Indexed: 05/22/2025]
Abstract
Microglial phagocytosis genes have been linked to increased risk for Alzheimer's disease (AD), but the mechanisms translating genetic association to cellular dysfunction remain unknown. Here, we showed that microglia formed lipid droplets (LDs) upon amyloid-β (Aβ) exposure and that LD loads increased with proximity to amyloid plaques in brains from individuals with AD and the 5xFAD mouse model. LD-laden microglia exhibited defects in Aβ phagocytosis, and unbiased lipidomic analyses identified a parallel decrease in free fatty acids (FFAs) and increase in triacylglycerols (TGs) as the key metabolic transition underlying LD formation. Diacylglycerol O-acyltransferase 2 (DGAT2)-a key enzyme that converts FFAs to TGs-promoted microglial LD formation and was increased in mouse 5xFAD and human AD brains. Pharmacologically targeting DGAT2 improved microglial uptake of Aβ and reduced plaque load and neuronal damage in 5xFAD mice. These findings identify a lipid-mediated mechanism underlying microglial dysfunction that could become a therapeutic target for AD.
Collapse
Affiliation(s)
- Priya Prakash
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Palak Manchanda
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Evi Paouri
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Kanchan Bisht
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Kaushik Sharma
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Jitika Rajpoot
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Victoria Wendt
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Ahad Hossain
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | | | - Caitlin E Randolph
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Yihao Chen
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Sarah Stanko
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nadia Gasmi
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Anxhela Gjojdeshi
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Sophie Card
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Jonathan Fine
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Krupal P Jethava
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Matthew G Clark
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Bin Dong
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Seohee Ma
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Alexis Crockett
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Elizabeth A Thayer
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Marlo Nicolas
- Division of Pathology, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Ryann Davis
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Dhruv Hardikar
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Daniela Allende
- Division of Pathology, Cleveland Clinic, Cleveland, OH 44106, USA
| | | | - Chi Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Dimitrios Davalos
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Gaurav Chopra
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
7
|
Sola-Sevilla N, Garmendia-Berges M, Aleixo M, Mera-Delgado MC, Solas M, Tordera RM, García-Carracedo L, Expósito S, Anaya-Cubero E, Fernández-Irigoyen J, Guruceaga E, Santamaria E, Martin ED, Puerta E. Microglial SIRT2 deficiency aggravates cognitive decline and amyloid pathology in Alzheimer's disease. Brain Behav Immun 2025; 129:223-243. [PMID: 40499846 DOI: 10.1016/j.bbi.2025.06.016] [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: 12/21/2024] [Revised: 05/17/2025] [Accepted: 06/07/2025] [Indexed: 06/16/2025] Open
Abstract
Sirtuin 2 (SIRT2), a NAD+-dependent deacetylase, has been implicated in aging and neurodegenerative diseases such as Alzheimer's disease (AD). While global SIRT2 inhibition has shown promise in reducing amyloid-beta pathology and cognitive deficits in different mouse models of AD, peripheral SIRT2 inhibition has been associated with adverse effects, such as increased inflammation. This suggests that targeted inhibition of specific cellular populations within the brain may represent a more precise and effective approach for the treatment of AD. To explore this hypothesis, we generated a conditional microglial SIRT2 knockout mouse model in the context of AD. Our results reveal that microglial SIRT2 reduction does not confer protective effects in the APP/PS1 model; rather, it aggravates cognitive decline, accelerates amyloid plaque deposition, and increases levels of pro-inflammatory cytokines at early stages of AD pathology. Transcriptomic analysis further indicates that SIRT2-deficient microglia exhibit altered expression of genes associated with aging and synaptic dysfunction. This phenotype was accompanied by increased phagocytosis of PSD95 and impaired long-term potentiation. These findings suggest that while SIRT2 inhibition in some contexts may be beneficial, targeted inhibition within microglia could accelerate AD progression, underscoring the need for cell-specific approaches when considering SIRT2 as a therapeutic target.
Collapse
Affiliation(s)
- Noemi Sola-Sevilla
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain; IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain; Department of Neurology, Research Center for Immunotherapy (FZI) and Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Centre of the Johannes Gutenberg University Mainz 55131 Mainz, Germany
| | - Maider Garmendia-Berges
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain; IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Mikel Aleixo
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain; IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - MCarmen Mera-Delgado
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain; IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Maite Solas
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain; IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Rosa M Tordera
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain; IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain
| | - Lucía García-Carracedo
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Sara Expósito
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Elena Anaya-Cubero
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
| | - Elisabeth Guruceaga
- IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain; Bioinformatics Platform, CIMA, University of Navarra, Pamplona, Spain
| | - Enrique Santamaria
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
| | - Eduardo D Martin
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Elena Puerta
- Department of Pharmaceutical Sciences, Division of Pharmacology, University of Navarra, 31008 Pamplona, Spain; IdISNA, Navarra Institute for Health Research, 31008 Pamplona, Spain.
| |
Collapse
|
8
|
Kordi R, Andrews TJ, Hicar MD. Infections, genetics, and Alzheimer's disease: Exploring the pathogenic factors for innovative therapies. Virology 2025; 607:110523. [PMID: 40174330 DOI: 10.1016/j.virol.2025.110523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative condition that creates a significant global health challenge and profoundly affects patients and their families. Recent research has highlighted the critical role of microorganisms, particularly viral infections, in the pathogenesis of AD. The involvement of viral infections in AD pathogenesis is predominantly attributed to their ability to induce neuroinflammation and amyloid beta (Aβ) deposition in the brain. The extant research exploring the relationship between viruses and AD has focused largely on Herpesviridae family. Traces of Herpesviruses, such as Herpes Simplex Virus-1 and Epstein Barr Virus, have been found in the brains of patients with AD. These viruses are thought to contribute to the disease progression by triggering chronic inflammatory responses in the brain. They can remain dormant in the brain, and become reactivated due to stress, a secondary viral infection, or immune-senescence in older adults. This review focuses on the association between Herpesviridae and bacterial infections with AD. We explore the genetic factors that might regulate viral illness and discuss clinical trials investigating antiviral and anti-inflammatory agents as possible therapeutic strategies to mitigate cognitive decline in patients with AD. In summary, understanding the interplay between infections, genetic factors, and AD pathogenesis may pave the way for novel therapeutic approaches, facilitating better management and possibly even prevent this debilitating disease.
Collapse
Affiliation(s)
- Ramesh Kordi
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Ted J Andrews
- Department of Pediatrics, Division of Developmental Pediatrics and Rehabilitation, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Mark D Hicar
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
9
|
Ayyubova G, Madhu LN. Microglial NLRP3 Inflammasomes in Alzheimer's Disease Pathogenesis: From Interaction with Autophagy/Mitophagy to Therapeutics. Mol Neurobiol 2025; 62:7124-7143. [PMID: 39951189 DOI: 10.1007/s12035-025-04758-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 02/08/2025] [Indexed: 05/15/2025]
Abstract
The nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome, discovered 20 years ago, is crucial in controlling innate immune reactions in Alzheimer's disease (AD). By initiating the release of inflammatory molecules (including caspases, IL-1β, and IL-18), the excessively activated inflammasome complex in microglia leads to chronic inflammation and neuronal death, resulting in the progression of cognitive deficiencies. Even though the involvement of NLRP3 has been implicated in neuroinflammation and widely explored in several studies, there are plenty of controversies regarding its precise roles and activation mechanisms in AD. Another prominent feature of AD is impairment in microglial autophagy, which can be either the cause or the consequence of NLRP3 activation and contributes to the aggregation of misfolded proteins and aberrant chronic inflammatory state seen in the disease course. Studies also demonstrate that intracellular buildup of dysfunctional and damaged mitochondria due to defective mitophagy enhances inflammasome activation, further suggesting that restoration of impaired autophagy and mitophagy can effectively suppress it, thereby reducing inflammation and protecting microglia and neurons. This review is primarily focused on the role of NLRP3 inflammasome in the etiopathology of AD, its interactions with microglial autophagy/mitophagy, and the latest developments in NLRP3 inflammasome-targeted therapeutic interventions being implicated for AD treatment.
Collapse
Affiliation(s)
- Gunel Ayyubova
- Department of Cytology, Embryology and Histology, Azerbaijan Medical University, Baku, Azerbaijan.
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M Health Science Center School of Medicine, College Station, TX, USA
| |
Collapse
|
10
|
Wang Y, Huang G, Liang X, Andrikopoulos N, Tang H, Ding F, Ke PC, Li Y. Microglial Clearance of Alzheimer's Amyloid-Beta Obstructed by Nanoplastics. ENVIRONMENTAL SCIENCE. NANO 2025; 12:3247-3260. [PMID: 40520627 PMCID: PMC12162090 DOI: 10.1039/d5en00291e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/18/2025]
Abstract
Understanding the neurological impact of nanoplastic exposure has become an area of intensive research recently. This study examined the molecular and cellular mechanisms of how nanoplastics affect amyloid-beta (Aβ) clearance by microglia in the context of Alzheimer's disease (AD). Transmission electron microscopy and molecular dynamics simulations showed that polystyrene nanoplastics accelerated Aβ aggregation by forming a protein corona, promoting peptide fibrillization through hydrogen bonding and π-π interactions. Flow cytometry and endocytosis inhibition assays revealed that polystyrene nanoplastics impaired microglial uptake of Aβ while increasing their own cellular internalization, leading to microglial energy depletion and allowing Aβ aggregates to evade immune clearance. Additionally, proteomic analysis indicated that polystyrene nanoplastics disrupted microglial homeostasis, exacerbated neuroinflammation and metabolic dysregulation, and impaired the signalling pathway of ABC transporters critical for Aβ clearance in the AD brain. These findings suggest that nanoplastics contribute to AD pathology by impeding Aβ clearance and corrupting neuroimmune defense.
Collapse
Affiliation(s)
- Yue Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| | - Gangtong Huang
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Xiufang Liang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| | - Nicholas Andrikopoulos
- Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Huayuan Tang
- Department of Engineering Mechanics, Hohai University, Nanjing 211100, China
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Pu Chun Ke
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Yuhuan Li
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China
| |
Collapse
|
11
|
Kokkali M, Karali K, Thanou E, Papadopoulou MA, Zota I, Tsimpolis A, Efstathopoulos P, Calogeropoulou T, Li KW, Sidiropoulou K, Gravanis A, Charalampopoulos I. Multimodal beneficial effects of BNN27, a nerve growth factor synthetic mimetic, in the 5xFAD mouse model of Alzheimer's disease. Mol Psychiatry 2025; 30:2265-2283. [PMID: 39587294 PMCID: PMC12092300 DOI: 10.1038/s41380-024-02833-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024]
Abstract
Alzheimer's Disease (AD) is an incurable and debilitating progressive, neurodegenerative disorder which is the leading cause of dementia worldwide. Neuropathologically, AD is characterized by the accumulation of Aβ amyloid plaques in the microenvironment of brain cells and neurovascular walls, chronic neuroinflammation, resulting in neuronal and synaptic loss, myelin and axonal failure, as well as significant reduction in adult hippocampal neurogenesis. The hippocampal formation is particularly vulnerable to this degenerative process, due to early dysfunction of the cholinergic circuit. Neurotrophic factors consist major regulatory molecules and their decline in AD is considered as an important cause of disease onset and progression. Novel pharmacological approaches are targeting the downstream pathways controlled by neurotrophins, such as nerve growth factor (NGF) receptors, TrkA and p75NTR, which enhance hippocampal neurogenic capacity and neuroprotective mechanisms, and potentially counteract the neurotoxic effects of amyloid deposition. BNN27 is a non-toxic, newly developed 17-spiro-steroid analog, penetrating the blood-brain-barrier (BBB) and mimicking the neuroprotective effects of NGF, acting as selective activator of its receptors, both TrkA and p75NTR, thus promoting survival of various neuronal cell types. Our present research aims at determining whether and which aspects of the AD-related pathology, BNN27 is able to alleviate, exploring the cellular and molecular AD components and link these changes with improvements in the cognitive performance of an animal AD model, the 5xFAD mice. Our results clearly indicate that BNN27 administration significantly reduced amyloid-β load in whole brain of the animals, enhanced adult hippocampal neurogenesis, restored cholinergic function and synaptogenesis, reducing inflammatory activation and leading to significant restoration of cognitive functions. BNN27 may represent a new lead multimodal molecule with neuroprotective, neurogenic and anti-neuroinflammatory actions for developing druggable anti-Alzheimeric agents. Proteomics data are available via ProteomeXchange with the identifier PXD044699.
Collapse
Affiliation(s)
- Maria Kokkali
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Kanelina Karali
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Evangelia Thanou
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics & Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Maria Anna Papadopoulou
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Ioanna Zota
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Alexandros Tsimpolis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | | | | | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics & Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Kyriaki Sidiropoulou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
- Department of Biology, School of Sciences and Engineering, University of Crete, Heraklion, 71003, Greece
| | - Achille Gravanis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, 71003, Greece.
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 71003, Greece.
| |
Collapse
|
12
|
Yang X, Li Y, Wei Z. Histogram-based analysis for confocal microscope images with immunofluorescent staining: A graphical-user-interface tool. MethodsX 2025; 14:103306. [PMID: 40271047 PMCID: PMC12017866 DOI: 10.1016/j.mex.2025.103306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
Immunofluorescent staining is widely utilized in biomedical research. However, reliable and reproducible quantification remains challenging and often depend on the experience of the raters heavily. As a result, variations caused by different raters or the same rater at different times confound quantitative interpretations. In this study, we propose a histogram-based analytical method to explore the sources of signals at various intensities and, subsequently, identify the signals of interest using the histograms as a reference. Our method aims to alleviate inter-rater and intra-rater inconsistencies and improve the quantification of microscope images with immunofluorescent staining.•Identification of signal sources for image pixels at different intensities•Reduction of quantification variations using the proposed histogram-based analysis.
Collapse
Affiliation(s)
- Xiuli Yang
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, United States
| | - Yuguo Li
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, United States
| | - Zhiliang Wei
- Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, United States
| |
Collapse
|
13
|
da Silva EA, Faber J, Penitente AR, Fernandes J, Bertolucci PHF, Longo BM, Arida RM. Effects of resistance exercise on behavioral and molecular changes in transgenic female mice for Alzheimer's disease in early and advanced stages. Exp Neurol 2025; 388:115217. [PMID: 40089002 DOI: 10.1016/j.expneurol.2025.115217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that affects memory and cognition, with a higher prevalence in women. Given the lack of effective treatment, physical activity stands out as a complementary approach to prevent or delay disease progression. While numerous studies on humans and animals indicate that aerobic exercise induces brain changes, the impact of resistance exercise (RE) on AD is not fully understood. OBJECTIVE This study aimed to investigate the behavioral and molecular changes induced by RE in female transgenic mice with AD at the early and advanced stages of the disease. MATERIALS AND METHODS Adult (initial phase - 7 to 8 months of age, n = 32) and adult/elderly (advanced phase - 22 to 23 months of age, n = 32) female mice (2xTg-AD) for the APPSWE/PS1dE9 mutation were subjected to a four-week RE protocol. Mobility, anxiety-like behavior, long-term memory (LTM), and depressive-like behavior were assessed. Beta-amyloid (βA) and cytokines were quantified using the ELISA technique. RESULTS There was a progressive increase in strength in both trained groups at different ages. RE reversed memory deficits only in adult AD animals and the anxiety-like behavior only in adult/elderly AD animals. RE reversed depressive-like behavior in adult and adult/elderly AD animals. RE reduced βA only in adult AD animals. RE modified the expression of several cytokines in animals in the early and advanced stage of AD. CONCLUSION RE can be a promising strategy to minimize the deleterious effects of AD; however, its effectiveness may be more limited to the early stages of the disease.
Collapse
Affiliation(s)
| | - Jean Faber
- Federal University of Sao Paulo - Neurology and Neurosurgery Department, Brazil
| | | | | | | | | | | |
Collapse
|
14
|
Ghanbarian E, Khorsand B, Petersen KK, Nallapu BT, Sajjadi SA, Lipton RB, Ezzati A, Alzheimer's Disease Neuroimaging Initiative *. Cerebrospinal fluid inflammatory cytokines as prognostic indicators for cognitive decline across Alzheimer's disease spectrum. J Alzheimers Dis 2025; 105:1298-1308. [PMID: 40262111 DOI: 10.1177/13872877251335915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
BackgroundNeuroinflammation actively contributes to the pathophysiology of Alzheimer's disease (AD); however, the value of neuroinflammatory biomarkers for disease-staging or predicting disease progression remains unclear.ObjectiveTo investigate diagnostic and prognostic utility of inflammatory biomarkers in combination with conventional AD biomarkers.MethodsData from 258 participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI) with cerebrospinal fluid (CSF) biomarkers of amyloid-β (Aβ), tau, and inflammation were analyzed. Clinically meaningful cognitive decline (CMCD) was defined as a ≥ 4-point increase on the Alzheimer's Disease Assessment Scale Cognitive Subscore 11. Predictor variables included demographics (D: age, sex, education), APOE4 status (A), inflammatory biomarkers (I), and classic AD biomarkers of Aβ and p-tau181 (C). Models incorporating inflammatory biomarkers assessed their contribution to improving baseline diagnostic classification and 1-year CMCD prediction.ResultsAt 1-year follow-up, 27.1% of participants experienced CMCD. Adding inflammatory biomarkers to models with D and A variables (DA model) improved classification of cognitively normal (CN) versus mild cognitive impairment (MCI) and CN versus Dementia (p < 0.001). Similarly, inflammatory markers enhanced classification in models including C (DAC model), for CN versus MCI (p < 0.01) and CN versus Dementia (p < 0.001). Predictive performance for CMCD was improved in individuals with MCI and dementia in both models (all p < 0.05). In addition, the DAI model outperformed the DAC model in predicting CMCD for MCI and Dementia groups (both p < 0.05).ConclusionsAddition of CSF inflammatory biomarkers to biomarkers of AD improves diagnostic accuracy of clinical disease stage at baseline and add incremental value to AD biomarkers for prediction of cognitive decline.
Collapse
Affiliation(s)
- Elham Ghanbarian
- Department of Neurology, University of California, Irvine, CA, USA
| | - Babak Khorsand
- Department of Neurology, University of California, Irvine, CA, USA
| | - Kellen K Petersen
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Bhargav T Nallapu
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - S Ahmad Sajjadi
- Department of Neurology, University of California, Irvine, CA, USA
| | - Richard B Lipton
- Saul B. Korey Department of Neurology, Albert Einstein College of Medicine, New York City, NY, USA
| | - Ali Ezzati
- Department of Neurology, University of California, Irvine, CA, USA
| | | |
Collapse
|
15
|
Samokhina E, Mangat A, Malladi CS, Gyengesi E, Morley JW, Buskila Y. Potassium homeostasis during disease progression of Alzheimer's disease. J Physiol 2025; 603:3405-3424. [PMID: 40366190 DOI: 10.1113/jp287903] [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: 10/18/2024] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease (AD) is an age-dependent neurodegenerative disorder characterized by neuronal loss leading to dementia and ultimately death. Whilst the loss of neurons is central to this disease, it is becoming clear that glia, specifically astrocytes, contribute to the onset and progression of neurodegeneration. The role of astrocytes in maintaining ion homeostasis in the extracellular milieu is fundamental for multiple brain functions, including synaptic plasticity and neuronal excitability, which are compromised during AD and affect neuronal signalling. In this study, we measured the astrocytic K+ clearance rate in the hippocampus and somatosensory cortex of a mouse model for AD during disease progression. Our results establish that astrocytic [K+]o (extracellular K+ concentration) clearance in the hippocampus is reduced in symptomatic 5xFAD mice, and this decrease is region-specific, as no significant alterations were detected in the superficial layers of the somatosensory cortex. The decrease in the [K+]o clearance rate correlated with a significant reduction in the expression and conductivity of Kir4.1 channels and a decline in the number of primary connected astrocytes. Moreover, astrocytes in the hippocampus of symptomatic 5xFAD mice demonstrated increased reactivity which was accompanied by an increased excitability and altered spiking profile of nearby neurons. These findings indicate that the supportive function astrocytes typically provide to nearby neurons is diminished during disease progression, which affects the neuronal circuit signalling in this area and provides a potential explanation for the increased vulnerability of neurons in AD. KEY POINTS: Astrocytic potassium clearance from the extracellular milleu is fundamental for multiple brain functions. Alterations in the clearance rate can affect the excitability and overall viability of neurons. A symptomatic mouse model for Alzheimer's disease (5xFAD) exhibits a significant decline in astrocytic K+ clearance at the hippocampus, but not the somatosensory cortex. The decrease in the clearance rate correlated with a reduction in the expression and conductivity of astrocytic Kir4.1 channels and a decrease in the number of primary connected astrocytes, specifically at the stratum lacunosum moleculare layer of the CA1 region. Astrocytes in the hippocampus of symptomatic 5xFAD mice displayed increased reactivity. The excitability profile and firing patterns of neurons at the hippocampus were affected by alterations in K+ homeostasis, indicating that the supportive function astrocytes typically provide to nearby neurons is diminished during progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Evgeniia Samokhina
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Armaan Mangat
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Chandra S Malladi
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Erika Gyengesi
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - John W Morley
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
- The MARCS Institute, Western Sydney University, Penrith, New South Wales, Australia
| |
Collapse
|
16
|
Skawratananond S, Xiong DX, Zhang C, Tonk S, Pinili A, Delacruz B, Pham P, Smith SC, Navab R, Reddy PH. Mitophagy in Alzheimer's disease and other metabolic disorders: A focus on mitochondrial-targeted therapeutics. Ageing Res Rev 2025; 108:102732. [PMID: 40122398 DOI: 10.1016/j.arr.2025.102732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/19/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Mitochondria, as central regulators of cellular processes such as energy production, apoptosis, and metabolic homeostasis, are essential to cellular function and health. The maintenance of mitochondrial integrity, especially through mitophagy-the selective removal of impaired mitochondria-is crucial for cellular homeostasis. Dysregulation of mitochondrial function, dynamics, and biogenesis is linked to neurodegenerative and metabolic diseases, notably Alzheimer's disease (AD), which is increasingly recognized as a metabolic disorder due to its shared pathophysiologic features: insulin resistance, oxidative stress, and chronic inflammation. In this review, we highlight recent advancements in pharmacological interventions, focusing on agents that modulate mitophagy, mitochondrial uncouplers that reduce oxidative phosphorylation, compounds that directly scavenge reactive oxygen species to alleviate oxidative stress, and molecules that ameliorate amyloid beta plaque accumulation and phosphorylated tau pathology. Additionally, we explore dietary and lifestyle interventions-MIND and ketogenic diets, caloric restriction, physical activity, hormone modulation, and stress management-that complement pharmacological approaches and support mitochondrial health. Our review underscores mitochondria's central role in the pathogenesis and potential treatment of neurodegenerative and metabolic diseases, particularly AD. By advocating for an integrated therapeutic model that combines pharmacological and lifestyle interventions, we propose a comprehensive approach aimed at mitigating mitochondrial dysfunction and improving clinical outcomes in these complex, interrelated diseases.
Collapse
Affiliation(s)
- Shadt Skawratananond
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Daniel X Xiong
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, United States.
| | - Charlie Zhang
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Sahil Tonk
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Aljon Pinili
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Brad Delacruz
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Patrick Pham
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Shane C Smith
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Rahul Navab
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, United States; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
17
|
Seplovich G, Bouchi Y, de Rivero Vaccari JP, Pareja JCM, Reisner A, Blackwell L, Mechref Y, Wang KK, Tyndall JA, Tharakan B, Kobeissy F. Inflammasome links traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Neural Regen Res 2025; 20:1644-1664. [PMID: 39104096 PMCID: PMC11688549 DOI: 10.4103/nrr.nrr-d-24-00107] [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: 01/25/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 08/07/2024] Open
Abstract
Traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease are three distinct neurological disorders that share common pathophysiological mechanisms involving neuroinflammation. One sequela of neuroinflammation includes the pathologic hyperphosphorylation of tau protein, an endogenous microtubule-associated protein that protects the integrity of neuronal cytoskeletons. Tau hyperphosphorylation results in protein misfolding and subsequent accumulation of tau tangles forming neurotoxic aggregates. These misfolded proteins are characteristic of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease and can lead to downstream neuroinflammatory processes, including assembly and activation of the inflammasome complex. Inflammasomes refer to a family of multimeric protein units that, upon activation, release a cascade of signaling molecules resulting in caspase-induced cell death and inflammation mediated by the release of interleukin-1β cytokine. One specific inflammasome, the NOD-like receptor protein 3, has been proposed to be a key regulator of tau phosphorylation where it has been shown that prolonged NOD-like receptor protein 3 activation acts as a causal factor in pathological tau accumulation and spreading. This review begins by describing the epidemiology and pathophysiology of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Next, we highlight neuroinflammation as an overriding theme and discuss the role of the NOD-like receptor protein 3 inflammasome in the formation of tau deposits and how such tauopathic entities spread throughout the brain. We then propose a novel framework linking traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease as inflammasome-dependent pathologies that exist along a temporal continuum. Finally, we discuss potential therapeutic targets that may intercept this pathway and ultimately minimize long-term neurological decline.
Collapse
Affiliation(s)
| | - Yazan Bouchi
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jennifer C. Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew Reisner
- Department of Pediatrics, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Laura Blackwell
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
18
|
Zhao JY, Zhou Y, Zhou CW, Zhan KB, Yang M, Wen M, Zhu LQ. Revisiting the critical roles of reactive microglia in traumatic brain injury. Int J Surg 2025; 111:3942-3978. [PMID: 40358653 PMCID: PMC12165506 DOI: 10.1097/js9.0000000000002420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/11/2025] [Indexed: 05/15/2025]
Abstract
Traumatic brain injury (TBI) triggers a complex neuroinflammatory cascade, with microglia serving as key regulators of both pathological damage and tissue structural restoration. Despite extensive research, the precise temporal evolution of microglial activation and its implications for long-term neurological outcomes remain incompletely understood. Here, we provide a comprehensive review of the molecular and cellular mechanisms underlying microglial responses in TBI, highlighting their role in neuroinflammation, neurogenesis, and tissue remodeling. We systematically compare clinical and preclinical TBI classifications, lesion patterns, and animal modeling strategies, evaluating their translational relevance. Furthermore, we explore the limitations of the conventional M1/M2 dichotomy and emphasize recent insights from single-cell transcriptomic analyses that reveal distinct microglial subpopulations across different injury phases. Finally, we discuss current therapeutic strategies targeting microglial modulation and propose future directions for neuroimmune interventions in TBI. By integrating findings from experimental and clinical studies, this review aims to bridge mechanistic insights with therapeutic advancements, paving the way for precision-targeted neuroimmune therapies.
Collapse
Affiliation(s)
- Jing-Yu Zhao
- Department of Neurosurgery, Wuhan Hankou Hospital, Hankou Hospital Affiliated to Wuhan University of Science and Technology, Jiang`an District, Wuhan, People’s Republic of China
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yang Zhou
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Chao-Wen Zhou
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ke-Bin Zhan
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, People’s Republic of China
| | - Ming Yang
- Department of Neurosurgery, Wuhan Hankou Hospital, Hankou Hospital Affiliated to Wuhan University of Science and Technology, Jiang`an District, Wuhan, People’s Republic of China
| | - Ming Wen
- Department of Neurosurgery, Wuhan Hankou Hospital, Hankou Hospital Affiliated to Wuhan University of Science and Technology, Jiang`an District, Wuhan, People’s Republic of China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
19
|
Zhang W, Teng F, Lan X, Liu P, Wang A, Zhang F, Cui Z, Guan J, Sun X. A novel finding relates to the involvement of ATF3/DOCK8 in Alzheimer's disease pathogenesis. J Alzheimers Dis 2025; 105:1385-1399. [PMID: 40267290 DOI: 10.1177/13872877251336266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundThe involvement of microglia is likely to be pivotal in the pathogenesis of Alzheimer's disease (AD) by modulating the deposition of amyloid-β (Aβ) plaques. The deletion of Dedicator of cytokinesis 8 (DOCK8) has a protective effect in mouse with neurodegenerative diseases.ObjectiveTo explore the underlying mechanism of DOCK8 in AD.MethodsIn present study, we first the detected the expression of DOCK8 in the hippocampal tissue of APP/PS1 mice. Then, the expression of DOCK8 was knocked down in the hippocampal tissue of APP/PS1 mice, and the effects of DOCK8 down-regulation on cognitive function, the microglia migration around Aβ plaques, and the cell division cycle 42 (Cdc42)/p38 mitogen-activated protein kinase (MAPK) signaling pathway were detected. Next, the effects of DOCK8 knockdown on Aβ-induced migration and activation of BV-2 cells as well as the MAPK signaling pathway were detected. Finally, the transcriptional regulation of DOCK by transcription factor 3 (ATF3) was detected by a dual luciferase reporter assay.ResultsDOCK8 expression exerts a significant upregulation in the hippocampus of APP/PS1 mice. However, following the DOCK8 knockdown, there was a significant recovery in the results of the behavioral tests and a notable reduction in microglial expression. Moreover, the high expression of DOCK8 mediated by ATF3 successfully triggered the Cdc42/p38 MAPK signaling pathway, thereby enhancing the migration and recruitment of microglia towards senile plaques, accelerating the production of Aβ plaques.ConclusionsATF3-mediated high expression of DOCK8 accelerates the production of Aβ plaques, and participates in the pathogenesis of AD.
Collapse
Affiliation(s)
- Wenqiang Zhang
- Department of Neurology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, People's Republic of China
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Fei Teng
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xifa Lan
- Department of Neurology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, People's Republic of China
| | - Peihui Liu
- Neurointerventional Department, Huludao Central Hospital, Huludao, Liaoning, People's Republic of China
| | - Aiming Wang
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Fan Zhang
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Zhiqiang Cui
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
- School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jingwei Guan
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaohong Sun
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
- Science Experiment Center, China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
20
|
Heneka MT, Gauthier S, Chandekar SA, Hviid Hahn-Pedersen J, Bentsen MA, Zetterberg H. Neuroinflammatory fluid biomarkers in patients with Alzheimer's disease: a systematic literature review. Mol Psychiatry 2025; 30:2783-2798. [PMID: 40050444 PMCID: PMC12092255 DOI: 10.1038/s41380-025-02939-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/08/2025] [Accepted: 02/19/2025] [Indexed: 05/22/2025]
Abstract
INTRODUCTION Neuroinflammation is associated with both early and late stages of the pathophysiology of Alzheimer's disease (AD). Fluid biomarkers are gaining significance in clinical practice for diagnosis in presymptomatic stages, monitoring, and disease prognosis. This systematic literature review (SLR) aimed to identify fluid biomarkers for neuroinflammation related to clinical stages across the AD continuum and examined long-term outcomes associated with changes in biomarkers. METHODS The SLR was conducted per the Cochrane Handbook for Systematic Reviews of Interventions and Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. We used PubMed®, Embase®, and Cochrane Collaboration databases to search for articles in English (between 2012 and 2022) on AD or mild cognitive impairment due to AD, using "neuroinflammation" or other "immune" search strings. Two independent reviewers screened titles and examined data from full-text articles for the SLR. RESULTS After the initial screening, 54 studies were prioritized for data extraction based upon their relevance to the SLR research questions. Nine studies for YKL-40, seven studies for sTREM2, and 11 studies for GFAP examined the relationship between the neuroinflammatory biomarkers and the clinical stage of the disease. Nine longitudinal studies further explored the association of fluid biomarkers with long-term clinical outcomes of disease. Cerebrospinal fluid (CSF) levels of YKL-40 were elevated in patients with AD dementia, while CSF sTREM2 levels were more strongly associated with preclinical and early symptomatic stages of AD. Plasma GFAP levels remained consistently elevated both in patients with AD dementia and individuals in preclinical stages with β-amyloid pathology. Longitudinal changes in plasma GFAP appeared to be predictive of cognitive decline in patients over time. DISCUSSION Neuroinflammatory biomarkers are associated with AD progression. More longitudinal studies in the preclinical and MCI stages of AD are needed to validate fluid biomarkers for diagnosis, disease monitoring, and prognosis in clinical practice.
Collapse
Affiliation(s)
- Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, Université du Luxembourg, Belvaux, Luxembourg.
| | - Serge Gauthier
- AD and Related Disorders Research Unit, McGill Center for Studies in Aging, Departments of Neurology & Neurosurgery, Psychiatry, and Medicine at McGill, Montreal, QC, Canada
| | | | | | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
21
|
Qin F, Cao R, Bai X, Yuan J, Sun W, Zheng Y, Qi X, Zhao W, Liu B, Gao C. Listerin Alleviates Alzheimer's Disease through IRE1-mediated Decay of TLR4 mRNA. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e14956. [PMID: 40448625 DOI: 10.1002/advs.202414956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 05/16/2025] [Indexed: 06/02/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, accounting for ≈60-70% of all dementia cases worldwide. Microglial-mediated brain inflammation is thought to play key roles in AD progression. Clinical evidence and animal models have indicated that the ribosome-associated quality control (RQC) component Listerin is involved in the development of AD. How Listerin regulates the development and progression of AD is unknown. Here, it is demonstrated that Listerin can decrease brain inflammation and alleviate AD-related cognitive impairments. Microglial-specific knockout of Listerin exhibits deteriorative cognitive symptoms based on the extracellular Amyloid-β (Aβ) or Lipopolysaccharide (LPS) injection. Mechanistically, Listerin directly binds to Toll-like receptor 4 (TLR4) mRNA and facilitates the IRE1α-mediated cleavage and degradation of TLR4 mRNA, leading to the alleviation of TLR4-induced brain inflammation. Adenovirus-mediated overexpression of Listerin decelerates the disease progression in the mouse model of Aβ-mediated neurodegeneration. Thus, Listerin is an important suppressor of microglia-induced brain inflammation and may be a potential therapeutic target for AD treatment.
Collapse
Affiliation(s)
- Fei Qin
- Key Laboratory of Infection, Immunity and prevention of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Runyu Cao
- Key Laboratory of Infection, Immunity and prevention of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Xuemei Bai
- Key Laboratory of Infection, Immunity and prevention of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Jiahua Yuan
- Key Laboratory of Infection, Immunity and prevention of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Wanwei Sun
- Key Laboratory of Infection, Immunity and prevention of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Yi Zheng
- Key Laboratory of Infection, Immunity and prevention of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Xiaopeng Qi
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Wei Zhao
- Key Laboratory of Infection, Immunity and prevention of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Bingyu Liu
- Key Laboratory of Infection, Immunity and prevention of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Chengjiang Gao
- Key Laboratory of Infection, Immunity and prevention of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, P. R. China
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, P. R. China
| |
Collapse
|
22
|
Bouwman MMA, Frigerio I, Lin CP, Reijner N, van de Berg WDJ, Jonkman LE. Hippocampal subfields: volume, neuropathological vulnerability and cognitive decline in Alzheimer's and Parkinson's disease. Alzheimers Res Ther 2025; 17:121. [PMID: 40448161 PMCID: PMC12124080 DOI: 10.1186/s13195-025-01768-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/18/2025] [Indexed: 06/02/2025]
Abstract
BACKGROUND The hippocampus is highly affected in neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). The relationship between neuropathology and atrophy in hippocampal subfields is complex due to differences in the selective neuronal vulnerability to distinct protein aggregates that underlie cognitive impairment. The aim of the current study was to investigate the relation between hippocampal subfield volumes, neuropathological burden (amyloid-β, p-tau and α-synuclein) and cognitive performance in AD, PD and control brain donors, using a cross-disease and within-subject post-mortem in situ MRI and neuropathology approach. METHODS A total of 60 brain donors, including 14 non-neurological controls, 27 AD and 19 PD, underwent post-mortem in situ MRI. From 3D-T1 images hippocampal subfield and entorhinal cortex volumes were derived using FreeSurfer-based subfield segmentation. Hippocampal tissue was obtained at subsequent autopsy, fixed and immunostained for amyloid-β, p-tau and pSer129-αSyn. Immunoreactivity in hippocampal subfields was quantified as area% load using QuPath. Clinical Dementia Rating scores were extracted from the clinical files when available. RESULTS AD showed atrophy and increased p-tau, but not amyloid-β, burden in the CA1, subiculum and entorhinal cortex compared to controls, however MRI and neuropathology did not correlate. Controls and PD had similar hippocampal subfield volumes and pathology load. In PD, p-tau pathology, rather than pSer129-αSyn, was associated with lower total hippocampal volume (r=-0.68, p = 0.045), predominantly in PD with dementia (PDD) (r=-0.99, p = 0.013). Cross-disease, volume loss of the subiculum (r=-0.68, p = 0.001) and entorhinal cortex (r=-0.73, p = 0.004) strongly associated with cognitive impairment. Moreover, p-tau pathology had the strongest effect on subfield atrophy, most pronounced in the subiculum (β=-0.570, p < 0.001), but could only explain 22-44% of the volumetric variance. CONCLUSIONS Even though p-tau was the strongest predictor of hippocampal subfield atrophy, AD-pathology (p-tau and amyloid-β) only partially accounted for volumetric differences in hippocampal subfields, highlighting the significance of other pathologies or mechanisms. The increased sensitivity of subicular and entorhinal cortical atrophy compared to total hippocampal atrophy highlights the potential clinical value of incorporating hippocampal subfield atrophy in monitoring disease progression.
Collapse
Affiliation(s)
- Maud M A Bouwman
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, location VUmc, De Boelelaan 1118, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands.
| | - Irene Frigerio
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, location VUmc, De Boelelaan 1118, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands
| | - Chen-Pei Lin
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, location VUmc, De Boelelaan 1118, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands
| | - Niels Reijner
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, location VUmc, De Boelelaan 1118, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands
| | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, location VUmc, De Boelelaan 1118, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Laura E Jonkman
- Department of Anatomy and Neurosciences, Section Clinical Neuroanatomy and Biobanking, Amsterdam UMC, location VUmc, De Boelelaan 1118, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Qu Y, Ding M, Zhang M, Zheng L, Hu B, An H. Iridoid glycosides in kidney-tonifying Chinese medicinal herbs: Mechanisms and implications for Alzheimer's disease therapy. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119870. [PMID: 40288663 DOI: 10.1016/j.jep.2025.119870] [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: 08/15/2024] [Revised: 03/22/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is an incurable and irreversible type of dementia. Existing drugs cannot meet clinical needs; thus, developing new treatments is necessary. Traditional Chinese medicine (TCM) has been used in the prevention and treatment of AD. TCM holds the theory that "the kidney support brain function" and believes that dementia can be addressed from a kidney-based perspective. Kidney-tonifying herbs are a class of medicines that have the effect of tonifying the kidney and benefiting the brain. Some of these herbs have been shown to have anti-AD effects. Iridoid glycosides (IGs), which are important components of kidney-tonifying herbs, may have the potential to prevent and treat AD. However, their effects on AD have not yet been reviewed. AIM OF THE REVIEW This literature review provides a comprehensive summary of the potential of IGs in the prevention and treatment of AD. It also sets the foundation for future studies that will make the use of such drugs in clinical practice possible. MATERIAL AND METHODS Kidney-tonifying Chinese herbs were selected with reference to the Chinese Pharmacopoeia (2020 edition) and the textbook of Chinese Materia Medica (5th edition). Literature survey was conducted using PubMed, Web of Science, Google Scholar, and CNKI, with "Alzheimer's disease," "kidney-tonifying Chinese medicinal herbs," and "Iridoid Glycosides" as the primary keywords. RESULTS Kidney-tonifying herbal IGs include loganin, morroniside, verbenalin, cornuside, catalpol, rehmannioside A, geniposidic acid, and aucubin. These IGs have shown multiple pharmacological effects, including anti-AD effects. The effective mechanisms of IGs for AD treatment include anti-oxidative stress, inhibiting neuronal apoptosis, antagonizing amyloid neurotoxicity and tau protein hyperphosphorylation, regulating immune function, anti-inflammation, normalizing the function of the cholinergic nervous system, recuperating neurobiochemical, and regulating AD-related genes. Consequently, IGs can combat AD by modulating multiple targets and pathways. CONCLUSION Kidney-tonifying herbal IGs have great potential to combat AD.
Collapse
Affiliation(s)
- Yanjie Qu
- Department of Traditional Chinese Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Minrui Ding
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Mengxue Zhang
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Bing Hu
- Cancer Institute, Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Hongmei An
- Department of Science & Technology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
24
|
Dahlén AD, Roshanbin S, Aguilar X, Bucher NM, Lopes van den Broek S, Sehlin D, Syvänen S. PET imaging of TREM2 in amyloid-beta induced neuroinflammation. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07358-0. [PMID: 40434494 DOI: 10.1007/s00259-025-07358-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025]
Abstract
PURPOSE The triggering receptor expressed on myeloid cells 2 (TREM2) has become a promising target for biologics in both monitoring and treating neuroinflammation in Alzheimer's disease (AD). This study aimed to develop and compare bispecific anti-TREM2 antibodies featuring different transferrin receptor (TfR) binders to enhance brain delivery, identifying the most suitable format for in vivo PET imaging of TREM2 in transgenic AD mice. METHODS Three bispecific TREM2-antibody formats were produced and evaluated for their ability to cross the blood-brain barrier (BBB) via TfR-mediated transcytosis and bind TREM2. Blood concentration profiles up to 72 h post-injection (p.i.), and ex vivo brain uptake of iodine-125-labeled antibody constructs were quantified in AppNL-G-F and age-matched wild type (WT) mice using a γ-counter. The best-performing bispecific TREM2-antibody was radiolabeled with iodine-124 and used for in vivo PET imaging of brain TREM2 levels in AppNL-G-F mice at 72 h p.i. Brain TREM2 concentrations were subsequently quantified using ELISA. RESULTS The antibody format carrying two scFv8D3 TfR-binders (IgG-scFv2), demonstrated the highest brain concentrations of all tested bispecific constructs. This antibody also exhibited significantly higher brain concentrations in AppNL-G-F mice compared to WT mice at both 48 and 72 h p.i. This difference was further visualized and quantified through in vivo PET imaging. Moreover, brain concentrations of the antibody ligand correlated with elevated TREM2 levels in brain homogenates. CONCLUSION These findings highlight IgG-scFv2 as a promising radioligand for in vivo PET imaging of TREM2, advancing non-invasive neuroinflammation studies and supporting drug development for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Amelia D Dahlén
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Sahar Roshanbin
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Ximena Aguilar
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Nadja M Bucher
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Sara Lopes van den Broek
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Dag Sehlin
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Stina Syvänen
- Department of Public Health and Caring Sciences, Section of Molecular Geriatrics, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
25
|
Putri PHL, Alamudi SH, Dong X, Fu Y. Extracellular vesicles in age-related diseases: disease pathogenesis, intervention, and biomarker. Stem Cell Res Ther 2025; 16:263. [PMID: 40437603 PMCID: PMC12121224 DOI: 10.1186/s13287-025-04374-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 05/02/2025] [Indexed: 06/01/2025] Open
Abstract
Aging is a multifactorial biological process characterized by the irreversible accumulation of molecular damage, leading to an increased risk of age-related diseases. With the global prominent rise in aging populations, elucidating the mechanisms underlying the aging process and developing strategies to combat age-related diseases have become a pressing priority. Extracellular vesicles (EVs) have gained significant attention due to their role in intercellular communication. EVs are known for their ability to deliver biocargoes, such as miRNA, proteins, and lipids, implicating their involvement in disease pathogenesis and intervention. In this review article, we explore the dual role of EVs in age-related diseases: contributing to the pathogenesis of diseases by transferring deleterious molecules, while also offering therapeutic ability by transferring beneficial molecules. We also highlight the application of EVs as biomarkers for early diagnosis of age-related diseases, paving the way for early intervention and precision medicine. Additionally, we discuss how analysing the composition of EVs cargo can provide insights into disease progression. Finally, we address the challenges and future perspectives of EV-based-therapy in clinical translation, including standardization of EVs isolation methods and improving cargo specificity.
Collapse
Affiliation(s)
- Puan Haliza Lintang Putri
- BGI Research, Hangzhou, 310030, China
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, 16242, Indonesia
| | - Samira Husen Alamudi
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, 16242, Indonesia
| | - Xuan Dong
- BGI Research, Hangzhou, 310030, China
| | - Ying Fu
- BGI Research, Hangzhou, 310030, China.
- Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
26
|
Aksnes M. Sex Differences in Biofluid Biomarkers for Alzheimer's Disease. NEURODEGENER DIS 2025:1-11. [PMID: 40418910 DOI: 10.1159/000545717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/01/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD), the most common cause of dementia, affects twice as many women as men. Moreover, sex is increasingly recognised as an important factor for AD, influencing symptom presentation, progression, disease biology, and treatment responses. In parallel, AD biomarkers are becoming more accessible with the discovery of specific and accurate blood-based biomarkers and their incorporation in AD diagnostic frameworks. This narrative review aimed to summarise sex differences in the concentration and interpretation of biofluid biomarkers for AD. SUMMARY Biological sex may influence both the concentration and interpretation of biofluid biomarkers for AD pathology such as amyloid-β aggregation, tau neurofibrillary tangles, neurodegeneration, or neuroinflammation. While some biofluid biomarkers display consistent sex differences in absolute levels, most biomarker levels have not been found to differ consistently by sex. Nonetheless, even biomarkers that do not differ in absolute levels display sex-specific associations with clinically relevant variables such as brain atrophy, cognitive impairment, and disease progression. KEY MESSAGE Sex may influence the interpretation of AD biomarkers depending on their context of use, and more research is required to develop sex-specific guidelines. Future research should aim to study sex differences and sex-specific associations with variables of interest, as well as the underlying factors driving sex differences in AD.
Collapse
Affiliation(s)
- Mari Aksnes
- Department of Geriatric Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
27
|
Roy KK, Mehta DK, Das R. Reevaluating Alzheimer's disease treatment: Can phytochemicals bridge the therapeutic Gap? Neuroscience 2025; 575:1-18. [PMID: 40216186 DOI: 10.1016/j.neuroscience.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Alzheimer's disease (AD) is a growing neurological disorder giving impact cognition and memory, posing a global health challenge with over 55 million individuals affected. It is the 7th foremost cause of dying worldwide, and its pervasiveness is expected to twofold in each five years, reaching 115 million by 2050. AD is characterized by neurofibrillary tangles, senile plaques, and oxidative stress, leading to synaptic failure and cognitive decline. Currently, there is no cure, and available FDA-approved drugs provide only symptomatic relief. The disease progresses through five phases- mild cognitive impairment (MCI), very severe, severe, moderate and mild AD. Research on AD focuses on various neurodegenerative pathways, including inflammation, oxidative stress, genetic factors, environmental variables, and amyloid-beta accumulation. Existing FDA-accepted drugs, like rivastigmine, memantine, galantamine, and donepezil, primarily address early symptoms but have limitations, including side effects and high costs. In this context, phytochemicals from plants, such as resveratrol, huperzine, quercetin, galantamine, and rosmarinic acid, show promise as potential treatments for AD and overcome the challenges and limitation of conventional treatment. These natural substances are being investigated for their ability to lower the risk of AD safely. However, there is a lack of comprehensive knowledge about their application, necessitating further research and clinical trials to explore their potential benefits and limitations. This review serves as an essential reference for advancing future studies on Alzheimer's disease. By thoroughly analyzing neurodegenerative pathways, addressing drug limitations, and highlighting the potential of phytochemicals, we establish a strong foundation for developing innovative therapeutic strategies. Closing the knowledge gap related to the use of phytochemicals in Alzheimer's management is not just important; it is critical for creating novel and more effective treatments for this challenging neurological condition.
Collapse
Affiliation(s)
- Kishor Kumar Roy
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India
| | - Dinesh Kumar Mehta
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India
| | - Rina Das
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be) University, Mullana, Ambala, HR, India.
| |
Collapse
|
28
|
Peng M, Zeng Q, Zheng W, Xia X. Peripheral Choroid/RPE/Sclera as a Shared Pathogenic Hub: Multi-Tissue Transcriptomic Profiling Identifies Common Differentially Expressed Genes in Age-Related Macular Degeneration and Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-05078-y. [PMID: 40411685 DOI: 10.1007/s12035-025-05078-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/15/2025] [Indexed: 05/26/2025]
Abstract
BACKGROUND Age-related macular degeneration (AMD) and Alzheimer's disease (AD), two prevalent neurodegenerative disorders, share overlapping pathophysiological features yet lack cross-disease therapeutic strategies. This study systematically investigates their parallel genes and shared molecular mechanisms to identify potential therapeutic targets for dry AMD, a condition with limited treatment options. METHODS Transcriptomic datasets for AMD (GSE155154) and AD (GSE95587) were retrieved from the GEO database. AMD tissues were stratified into four subgroups: macular retina (MR), macular choroid/RPE/sclera (MCRS), peripheral retina (PR), and peripheral choroid/RPE/sclera (PCRS). Common differentially expressed genes (DEGs) were identified and analyzed via functional enrichment (GO, KEGG), gene set enrichment analysis (GSEA), and protein-protein interaction (PPI) networks. Drug-gene interactions and competing endogenous RNA (ceRNA) networks were constructed to prioritize therapeutic targets. Key hub genes were experimentally validated in a sodium iodate-induced AMD murine model using RT-qPCR. RESULTS Comparative analysis revealed 89, 56, 4, and 130 common DEGs between AD and MR, MCRS, PR, and PCRS subgroups, respectively. Neuroactive ligand-receptor interactions were prioritized in MR/MCRS-AD analyses, while extracellular matrix organization emerged as the dominant pathway in PCRS-AD comparisons. GSEA identified conserved the TNFα signaling pathway via NF-κB across both diseases. PCRS exhibited consistent expression trends for shared genes and pathways with AD. Computational screening prioritized seven druggable targets (COL1A1, COL1A2, COL3A1, MMP2, MMP9, VCAN, ITGA5) with dual therapeutic potential, along with a reconstructed circRNA (circRNA_002179)-miRNA (miR-124)-mRNA (VCAN) regulatory axis. Experimental validation in a sodium iodate-induced AMD murine model confirmed region-specific dysregulation: hub genes were significantly downregulated in MCRS but upregulated in PCRS. CONCLUSIONS Our study delineates both convergent and divergent molecular landscapes of AMD and AD, with PCRS emerging as a critical locus for shared pathophysiology. These findings bridge a critical gap in understanding AMD-AD comorbidity, offering actionable strategies for targeted drug development.
Collapse
Affiliation(s)
- Manjuan Peng
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Ophthalmology, Hunan Provincial People's Hospital and The First-Affiliated Hospital of Hunan Normal University, Changsha, China
- Department of Ophthalmology, the 921, Hospital of PLA (Second Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Qi Zeng
- Department of Ophthalmology, Hunan Provincial People's Hospital and The First-Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Wei Zheng
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
29
|
Mario A, Ivana L, Claudia MM, Antonello B, Francesco P, Tommaso C, Madia L. Can ketamine therapy overcome treatment-resistant depression in Alzheimer's disease and older adults? Preclinical and clinical evidence. Biomed Pharmacother 2025; 188:118199. [PMID: 40412361 DOI: 10.1016/j.biopha.2025.118199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 05/11/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025] Open
Abstract
Treatment-resistant depression (TRD) presents substantial clinical challenges, particularly in patients with Alzheimer's disease (AD) and older adults experiencing late-life depression. Traditional monoaminergic therapies often fail in this population due to neurodegenerative changes that impact receptor dynamics and neurotransmitter systems. Emerging evidence suggests that N-methyl-D-aspartate (NMDA) receptor antagonists, such as ketamine, esketamine, and arketamine, may offer new avenues for treatment. This review examines the potential of ketamine and its derivatives in treating TRD in older adults and individuals with AD, focusing on their mechanisms of action, clinical efficacy, and limitations in the context of neurodegenerative pathology. Following PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines, we conducted a systematic search of PubMed, Google Scholar, and Web of Science databases up until January 2025, with no year restrictions. Nineteen human clinical studies and eight preclinical studies met the inclusion criteria. Evidence suggests that ketamine may offer advantages over standard treatments for AD, potentially due to its broader mechanism of action compared to the NMDA antagonist memantine, as observed in animal models of AD. Clinical findings have demonstrated the rapid and robust antidepressant effects of ketamine and esketamine, alleviating depressive symptoms in both AD patients and older adults with TRD, indicating their potential as effective therapeutic options for these complex conditions.
Collapse
Affiliation(s)
- Altamura Mario
- Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy.
| | - Leccisotti Ivana
- Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | | | - Bellomo Antonello
- Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Panza Francesco
- Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro", Bari, Italy
| | - Cassano Tommaso
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Lozupone Madia
- Department of Translational Biomedicine and Neuroscience "DiBrain", University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
30
|
Liu X, Baxley S, Hebron M, Turner RS, Moussa C. Resveratrol Attenuates CSF Markers of Neurodegeneration and Neuroinflammation in Individuals with Alzheimer's Disease. Int J Mol Sci 2025; 26:5044. [PMID: 40507855 PMCID: PMC12155158 DOI: 10.3390/ijms26115044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2025] [Revised: 05/16/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by amyloid-beta (Aβ) accumulation and neuroinflammation. A previous multicenter, phase 2, double-blind, placebo-controlled trial randomized 179 participants into placebo or resveratrol over 52 weeks. Sub-analysis of CSF biomarkers of neuronal damage, inflammation, and microglial activity was performed in a subset of patients treated with a placebo (n = 21) versus resveratrol (n = 30). Markers of neuronal damage, including neuron-specific enolase and hyperphosphorylated neurofilaments, were reduced. Microglial activation was measured via a triggering receptor expressed on myeloid cells (TREM)-2 at baseline and after resveratrol treatment. Resveratrol significantly reduced CSF TREM2 levels and decreased inflammation and tissue damage, including matrix metalloprotease (MMP)-9. Cathepsin D, a lysosomal marker of autophagy, was reduced in the resveratrol group compared with placebo, while angiogenin, a marker of vascular angiogenesis, was increased. These data suggest that resveratrol may exert anti-inflammatory and neuroprotective effects in AD by reducing CSF TREM2 and other markers of neuronal damage. Further research is needed to assess the significance of these biomarker changes on clinical outcomes in patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA; (X.L.); (S.B.); (M.H.)
| | - Sean Baxley
- Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA; (X.L.); (S.B.); (M.H.)
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Michaeline Hebron
- Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA; (X.L.); (S.B.); (M.H.)
| | - Raymond Scott Turner
- Memory Disorders Program, Department of Neurology, Georgetown University, Washington, DC 20057, USA;
| | - Charbel Moussa
- Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC 20057, USA; (X.L.); (S.B.); (M.H.)
| |
Collapse
|
31
|
Bhogal I, Pankaj V, Roy S. Identifying RAGE inhibitors as potential therapeutics for Alzheimer's disease via integrated in-silico approaches. Sci Rep 2025; 15:17730. [PMID: 40404684 PMCID: PMC12098779 DOI: 10.1038/s41598-025-01271-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
Alzheimer's disease is a neurodegenerative disorder characterized by two hallmarks: amyloid beta plaques and neurofibrillary tangles. The receptor for advanced glycation end products (RAGE) is a multi-ligand receptor involved in the pathophysiology of various diseases including cancer, diabetes, cardiovascular diseases, and Alzheimer's disease (AD). Therefore, targeting RAGE could be an effective strategy to block RAGE signaling pathways. The present study aims to identify potential RAGE inhibitors against AD through comprehensive in-silico approaches. A total of 708,580 compounds were screened from numerous databases using structure-based virtual screening and ADMET evaluation. Further, the molecules with good glide scores were assessed by molecular docking studies. Subsequently, the top six ligands were subjected to molecular dynamic (MD) simulations for 100 ns and binding free energy calculations to check their stability with RAGE (PDB: 6XQ3). The per-residue decomposition analysis revealed that specific residues namely, GLY_20, ALA_21, LYS_39, GLU_50, LYS_52, ARG_98, GLN_100, LYS_110, ASN_112, and ARG_198 played a key role in the binding process. Furthermore, the trajectory analysis (DCCM and PCA) analyzed the dominant motions of residues and predicted the stability of protein-ligand complexes. In conclusion, the Hit-6 compound could be a promising candidate for targeting RAGE and deserves further consideration as an anti-Alzheimer drug.
Collapse
Affiliation(s)
- Inderjeet Bhogal
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, 616 00, Czech Republic
| | - Vaishali Pankaj
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, 616 00, Czech Republic
| | - Sudeep Roy
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, 616 00, Czech Republic.
| |
Collapse
|
32
|
Gu L, Mao X, Tian C, Yang Y, Yang K, Canfield SG, Zhu D, Gu M, Guo F. Engineering blood-brain barrier microphysiological systems to model Alzheimer's disease monocyte penetration and infiltration. Biomater Sci 2025. [PMID: 40391576 DOI: 10.1039/d5bm00204d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Alzheimer's disease (AD) is a progressive and neurodegenerative disease, predominantly causing dementia. Despite increasing clinical evidence suggesting the involvement of peripheral immune cells such as monocytes in AD pathology, the dynamic penetration and infiltration of monocytes crossing blood-brain barrier (BBB) and inducing neuroinflammation is largely understudied in an AD brain. Herein, we engineer BBB-like microphysiological system (BBB-MPS) models for recapitulating the dynamic penetration and infiltration of monocytes in an AD patient's brain. Each BBB-MPS model can be engineered by integrating a functional BBB-like structure on a human cortical organoid using a 3D-printed device within a well of a plate. By coculturing these BBB-MPS models with monocytes from AD patients and age-matched healthy donors, we found that AD monocytes exhibit significantly greater BBB penetration and brain infiltration compared to age-matched control monocytes. Moreover, we also tested the interventions including Minocycline and Bindarit, and found they can effectively inhibit AD monocyte infiltration, subsequently reducing neuroinflammation and neuronal apoptosis. We believe these scalable and user-friendly BBB-MPS models may hold promising potential in modeling and advancing therapeutics for neurodegenerative and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Xiangdi Mao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Chunhui Tian
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Yang Yang
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Kaiyuan Yang
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Scott G Canfield
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Terre Haute, IN, 47809, USA
| | - Donghui Zhu
- Department of Biomedical Engineering, University of Stony Brook, 100 Nicolls Rd, Stony Brook, NY, 11794, USA
| | - Mingxia Gu
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, University of California, Los Angeles, CA 90095, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
33
|
Clark R, Lira-Junior R, Jansson L, Eriksdotter M, Schultzberg M, Pussinen P, Buhlin K, Boström EA. Elevated levels of salivary interleukin-34 in patients suffering from Alzheimer's disease. Clin Oral Investig 2025; 29:303. [PMID: 40389754 PMCID: PMC12089168 DOI: 10.1007/s00784-025-06376-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/05/2025] [Indexed: 05/21/2025]
Abstract
OBJECTIVES To investigate interleukin (IL)-34 and colony-stimulating factor (CSF)-1 levels in saliva, cerebrospinal fluid, and plasma in different stages of cognitive impairment. The study also examines the relationship between these biomarkers and periodontal status across different stages of cognitive impairment. MATERIAL AND METHODS A total of 230 individuals diagnosed with Alzheimer's disease (AD, n = 52), mild cognitive impairment (MCI, n = 51), subjective cognitive impairment (SCI, n = 51), and controls (n = 76) were enrolled. Participants underwent clinical and radiological oral examinations. Cerebrospinal fluid samples were collected from all groups except controls. Stimulated saliva and blood were collected during oral examination. IL-34 and CSF-1 levels were assessed using enzyme-linked immunosorbent assays. RESULTS Salivary IL-34 levels were increased in AD compared to SCI (p = 0.010) and controls (p < 0.001), and in MCI compared to controls (p < 0.001). Elevated salivary CSF-1 levels were observed in AD compared to SCI (p = 0.003). Salivary IL-34 was inversely associated with Mini-Mental State Examination (MMSE) scores (p < 0.010) and body mass index (p = 0.040), while CSF-1 was associated with age (p = 0.015). IL-34 and CSF-1 levels did not differ in cerebrospinal fluid between groups, and periodontal status did not affect the levels in any biofluid measured. CONCLUSION Salivary IL-34 is increased in AD patients and is associated with MMSE scores. CLINICAL RELEVANCE Identifying reliable biomarkers for AD is crucial for early detection and intervention. This study suggests that salivary IL-34 could serve as a potential biomarker for AD.
Collapse
Affiliation(s)
- Reuben Clark
- Division of Oral diagnostics and surgery, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden.
| | - Ronaldo Lira-Junior
- Division of Oral diagnostics and surgery, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Leif Jansson
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Maria Eriksdotter
- Center for Alzheimer Research, Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Marianne Schultzberg
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Pirkko Pussinen
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
- Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
| | - Kåre Buhlin
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Elisabeth A Boström
- Division of Oral diagnostics and surgery, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
- Department of Orofacial Medicine, Folktandvården Stockholms Län AB, Stockholm, Sweden
| |
Collapse
|
34
|
Park Y, Moon S, Jung H, Park S, Kim JW, Song DG, In YH, Han SW, Sohn JH, Lee CH. Mirodenafil improves cognitive function by reducing microglial activation and blood-brain barrier permeability in ApoE4 KI mice. Front Aging Neurosci 2025; 17:1579411. [PMID: 40443793 PMCID: PMC12119498 DOI: 10.3389/fnagi.2025.1579411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/24/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Alzheimer's disease (AD) has significant public health concerns in the aging society. AD can compromise brain function and lead to severe neurological abnormalities associated with dementia. The human Apolipoprotein E (ApoE4) gene is a strong risk factor for AD. However, comprehensive analyses and improvements of mouse models expressing ApoE4 remain largely unexplored. Methods ApoE4 knock-in (KI) mice were used to investigate the role of humanized ApoE4 in hippocampal histological changes and cognitive impairment. Cerebrovascular perfusion, blood-brain barrier (BBB) integrity, microgliosis, and amyloid-beta 42 (Aβ42) accumulation were examined. Cognitive functions were assessed using the Morris water maze, Y-maze, and novel object recognition tests. Mirodenafil, a potent and selective phosphodiesterase 5 inhibitor (PDE5i), was orally administered to ApoE4 KI mice for 4 weeks. An in vitro BBB model and BV2 microglial cells were used to investigate endothelial permeability and inflammation. Results ApoE4 KI mice exhibited not only reduced cerebrovascular perfusion and CLN-5 expression but also increased microgliosis and Aβ42 accumulation in the hippocampus. These phenomena were accompanied by impaired cognitive functions. Mirodenafil administration reversed the histological and behavioral alterations induced by ApoE4 KI. In vitro, mirodenafil treatment mitigated Aβ42-induced endothelial permeability and lipopolysaccharide-induced microglial inflammation. Discussion These findings suggest that mirodenafil enhances cerebrovascular function, preserves BBB integrity, and mitigates neuroinflammation in ApoE4 KI mice, leading to cognitive improvement. PDE5 inhibition may serve as a promising therapeutic approach for addressing ApoE4-associated cerebrovascular and cognitive dysfunction.
Collapse
Affiliation(s)
- Yejin Park
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| | - Subin Moon
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| | - Harry Jung
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Songyi Park
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Ju Won Kim
- AriBio Co., Ltd., Seongnam-si, Gyeonggi-Do, Republic of Korea
| | - Dan-Gyeong Song
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| | - Yong-Ho In
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Sang Won Han
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon, Republic of Korea
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Jong-Hee Sohn
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon, Republic of Korea
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Chan Hee Lee
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
- Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
35
|
Battistella G, Xavier LDL, Vortmeyer AO, Simonyan K. Abnormal Brain Iron Metabolism is Linked to Altered Neural Function in Isolated Laryngeal Dystonia. Mov Disord 2025. [PMID: 40370031 DOI: 10.1002/mds.30217] [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: 01/27/2025] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Laryngeal dystonia (LD) is an isolated focal dystonia causing involuntary spasms in the laryngeal muscles that selectively impair speech production. LD is characterized as a functional and structural neural network disorder; however, the mechanistic aspects of network dysfunction in dystonia remain unknown. OBJECTIVE We hypothesized that iron-induced abnormal metabolic processes may underlie microstructural neuronal damage, contributing to altered neural activity within the dystonic network and, subsequently, the development of the dystonic state. METHODS We used 7 Tesla magnetic resonance imaging (MRI) at ultra-high field resolution for quantitative susceptibility mapping (QSM) of iron content, multi-echo multi-band resting-state functional MRI (fMRI) of brain activity and functional connectivity, positron emission tomography with [11C]flumazenil radioligand of GABAA neuroreceptor availability, and immunohistochemistry of postmortem brain tissue to investigate iron metabolism in LD patients and healthy controls. RESULTS The QSM analysis found increased iron content in primary sensorimotor and premotor cortices, inferior frontal, middle frontal, and middle temporal gyri, middle cingulate cortex, superior and inferior parietal lobules, insula, putamen, and cerebellum. Histopathology substantiated the neuroimaging findings by showing focal clusters of iron precipitates in these regions. Increased iron content in the supplementary motor area and middle cingulate cortex was associated with altered neural activity, while increased iron in the middle cingulate cortex, premotor cortex, and putamen had associations with GABAA receptor availability in LD patients. CONCLUSION Abnormal iron accumulations are likely to contribute to the imbalance of excitatory and inhibitory signaling within the dystonic neural network, leading to altered network dynamics that ultimately contribute to LD development. © 2025 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Giovanni Battistella
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, Massachusetts, USA
| | - Laura de Lima Xavier
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, Massachusetts, USA
| | - Alexander O Vortmeyer
- Department of Pathology and Laboratory Medicine, University of Indiana, Indianapolis, USA
| | - Kristina Simonyan
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear and Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
36
|
Gomes-da-Silva NC, Xavier-de-Britto I, Soares MAG, Yoshihara NMA, Ilem Özdemir D, Ricci-Junior E, Fechine PBA, Alencar LMR, Henriques MDGMDO, Barja-Fidalgo TC, Follmer C, Santos-Oliveira R. Nanostructured Lipoxin A4: Understanding Its Biological Behavior and Impact on Alzheimer's Disease (Proof of Concept). Pharmaceutics 2025; 17:649. [PMID: 40430939 PMCID: PMC12114923 DOI: 10.3390/pharmaceutics17050649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/09/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Lipoxins, particularly Lipoxin A4 (LXA4), are endogenous lipid mediators with potent anti-inflammatory and pro-resolving properties, making them promising candidates for the treatment of inflammatory and neurodegenerative disorders. However, their therapeutic application is limited by poor stability and bioavailability. This study aimed to develop and characterize nanomicelles encapsulating LXA4 (nano-lipoxin A4) to improve its pharmacological efficacy against Alzheimer's disease (AD), a neurodegenerative condition marked by chronic inflammation and beta-amyloid (Aβ) accumulation. Methods: Nano-lipoxin A4 was synthesized using Pluronic F-127 as a carrier and characterized in terms of morphology, physicochemical stability, and in vitro activity against Aβ fibrils. Dissociation of Aβ fibrils was assessed via Thioflavin-T fluorescence assays and transmission electron microscopy. In vivo biodistribution and pharmacokinetic profiles were evaluated using technetium-99m-labeled nano-lipoxin A4 in rodent models. Hepatic biochemical parameters were also measured to assess potential systemic effects. Results: In vitro studies demonstrated that nano-lipoxin A4 effectively dissociated Aβ fibrils at concentrations of 50 nM and 112 nM. Electron microscopy confirmed the disruption of fibrillar structures. In vivo imaging revealed predominant accumulation in the liver and spleen, consistent with reticuloendothelial system uptake. Pharmacokinetic analysis showed a prolonged half-life (63.95 h) and low clearance rate (0.001509 L/h), indicating sustained systemic presence. Biochemical assays revealed elevated liver enzyme levels, suggestive of increased hepatic metabolism or potential hepatotoxicity. Conclusions: Nano-lipoxin A4 exhibits significant therapeutic potential for Alzheimer's disease through effective modulation of Aβ pathology and favorable pharmacokinetic characteristics. However, the elevation in liver enzymes necessitates further investigation into systemic safety to support clinical translation.
Collapse
Affiliation(s)
- Natália Cristina Gomes-da-Silva
- Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Nuclear Engineering Institute, Brazilian Nuclear Energy Commission, Rio de Janeiro 21941906, RJ, Brazil; (N.C.G.-d.-S.); (I.X.-d.-B.); (M.A.G.S.); (N.M.A.Y.)
| | - Isabelle Xavier-de-Britto
- Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Nuclear Engineering Institute, Brazilian Nuclear Energy Commission, Rio de Janeiro 21941906, RJ, Brazil; (N.C.G.-d.-S.); (I.X.-d.-B.); (M.A.G.S.); (N.M.A.Y.)
| | - Marilia Amável Gomes Soares
- Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Nuclear Engineering Institute, Brazilian Nuclear Energy Commission, Rio de Janeiro 21941906, RJ, Brazil; (N.C.G.-d.-S.); (I.X.-d.-B.); (M.A.G.S.); (N.M.A.Y.)
| | - Natalia Mayumi Andrade Yoshihara
- Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Nuclear Engineering Institute, Brazilian Nuclear Energy Commission, Rio de Janeiro 21941906, RJ, Brazil; (N.C.G.-d.-S.); (I.X.-d.-B.); (M.A.G.S.); (N.M.A.Y.)
| | - Derya Ilem Özdemir
- Faculty of Pharmacy, Department of Radiopharmacy, Ege University, Bornova, Izmir 35040, Turkey;
| | - Eduardo Ricci-Junior
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro 21941900, RJ, Brazil;
| | - Pierre Basílio Almeida Fechine
- Group of Chemistry of Advanced Materials (GQMat), Department of Analytical Chemistry and Physical-Chemistry, Federal University of Ceará, Fortaleza 451970, CE, Brazil;
| | | | - Maria das Graças Muller de Oliveira Henriques
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551030, RJ, Brazil; (M.d.G.M.d.O.H.); (T.C.B.-F.)
| | - Thereza Christina Barja-Fidalgo
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551030, RJ, Brazil; (M.d.G.M.d.O.H.); (T.C.B.-F.)
| | - Cristian Follmer
- Laboratory of Biological Chemistry of Neurodegenerative Disorders, Department of Physical Chemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941909, RJ, Brazil;
| | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Nuclear Engineering Institute, Brazilian Nuclear Energy Commission, Rio de Janeiro 21941906, RJ, Brazil; (N.C.G.-d.-S.); (I.X.-d.-B.); (M.A.G.S.); (N.M.A.Y.)
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Rio de Janeiro State University, Rio de Janeiro 23070200, RJ, Brazil
| |
Collapse
|
37
|
Chen YH, Wang ZB, Liu XP, Mao ZQ. Cerebrospinal Fluid CCL25 as a Biomarker for Alzheimer's Disease: Associations with Pathology, Neurodegeneration, and Cognitive Decline. Mol Neurobiol 2025:10.1007/s12035-025-05007-z. [PMID: 40366557 DOI: 10.1007/s12035-025-05007-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
Neuroinflammation plays a crucial role in Alzheimer's disease (AD) pathogenesis. We investigated the relationship between cerebrospinal fluid (CSF) C-C chemokine ligand 25 (CCL25), an inflammatory regulator, and AD pathology and progression. We analyzed data on CSF CCL25, AD biomarkers (CSF β-amyloid [Aβ]42, phosphorylated tau [pTau]181, amyloid positron emission tomography [PET]), postmortem neuropathology, magnetic resonance imaging-based neurodegeneration, and cognitive function from 703 participants in the Alzheimer's Disease Neuroimaging Initiative cohort. We found that elevated CSF CCL25 levels were associated with cognitive impairment, abnormal Aβ and tau pathology, greater brain atrophy, and worse cognitive performance (all P < 0.05). Notably, CSF CCL25 exhibited nonlinear relationships with Aβ and tau pathology, reaching a plateau as AD pathology increased. CSF CCL25 showed acceptable diagnostic accuracy in distinguishing amyloid-positive/negative (A ±) and tau-positive/negative (T ±) participants (area under the curve [AUC] = 0.71-0.77) and autopsy-confirmed AD cases (AUC = 0.77), with optimal performance in differentiating A + T + from A-T- participants (AUC = 0.82-0.85 with age and sex adjustment). Longitudinally, higher baseline CSF CCL25 predicted accelerated amyloid accumulation, hippocampal atrophy, and cognitive decline. Mediation analyses revealed that CCL25 partially mediated associations between Aβ pathology and tau pathology (mediating effect: 54.5%), neurodegeneration (18.2%), and cognitive decline (7.4%). Among 37 CSF CCL and CXCL chemokines examined, 28 were associated with at least one AD-related outcome, with CCL25 demonstrating the strongest associations overall. These findings suggest that CSF CCL25 is involved in early AD pathological progression and may serve as an inflammatory biomarker for diagnosis and monitoring of disease progression in AD.
Collapse
Affiliation(s)
- Yu-Han Chen
- Department of Human Anatomy, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, 050017, China
- The First Clinical Medical School, Hebei North University, Zhangjiakou, 075000, China
| | - Zhi-Bo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Xi-Peng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Zhi-Qi Mao
- Department of Neurosurgery, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
38
|
Marongiu R, Platholi J, Park L, Yu F, Sommer G, Woods C, Milner TA, Glass MJ. Promotion of neuroinflammation in select hippocampal regions in a mouse model of perimenopausal Alzheimer's disease. Front Mol Biosci 2025; 12:1597130. [PMID: 40438709 PMCID: PMC12116374 DOI: 10.3389/fmolb.2025.1597130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/22/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Alzheimer's disease, the most common form of dementia, is characterized by age-dependent amyloid beta (Ab) aggregation and accumulation, neuroinflammation, and cognitive deficits. Significantly, there are prominent sex differences in the risk, onset, progression, and severity of AD, as well as response to therapies, with disease burden disproportionately affecting women. Although menopause onset (i.e., perimenopause) may be a critical transition stage for AD susceptibility in women, the role of early ovarian decline in initial disease pathology, particularly key neuroinflammatory processes, is not well understood. Methods To study this, we developed a unique mouse model of perimenopausal AD by combining an accelerated ovarian failure (AOF) model of menopause induced by 4-vinylcyclohexene diepoxide (VCD) with the 5xFAD transgenic AD mouse model. To target early stages of disease progression, 5xFAD females were studied at a young age (∼4 months) and at the beginning stage of ovarian failure analogous to human perimenopause (termed "peri-AOF"), and compared to age-matched males. Assessment of neuropathology was performed by immunohistochemical labeling of Ab as well as markers of astrocyte and microglia activity in the hippocampus, a brain region involved in learning and memory that is deleteriously impacted during AD. Results Our results show that genotype, AOF, and sex contributed to AD-like pathology. Aggregation of Ab was heightened in female 5xFAD mice and further increased at peri-AOF, with hippocampal subregion specificity. Further, select increases in glial activation also paralleled Ab pathology in distinct hippocampal subregions. However, cognitive function was not affected by peri-AOF. Discussion These findings align with the hypothesis that perimenopause constitutes a period of susceptibility for AD pathogenesis in women.
Collapse
Affiliation(s)
- Roberta Marongiu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
- Neurological Surgery Department, Weill Cornell Medicine, New York, NY, United States
- Genetic Medicine Department, Weill Cornell Medicine, New York, NY, United States
| | - Jimcy Platholi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
- Anesthesiology Department, Weill Cornell Medicine, New York, NY, United States
| | - Laibak Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Fangmin Yu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Garrett Sommer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Clara Woods
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Michael J. Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
39
|
Chen Y, Yang X, Li J, Luo H, Huang Q, Yang W, Lei T, Lui S, Gong Q, Li H, Wu H, Gao H. A nasally administrated reactive oxygen species-responsive carrier-free gene delivery nanosystem for Alzheimer's disease combination therapy. J Control Release 2025; 381:113604. [PMID: 40043915 DOI: 10.1016/j.jconrel.2025.113604] [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: 11/07/2024] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 03/10/2025]
Abstract
Combination therapies targeting multiple pathways are needed in order to improve treatment outcomes in Alzheimer's disease (AD) due to its complex pathogenesis. Amyloid-β and microglia-mediated neuroinflammation significantly contribute to AD pathogenesis. Amyloid-β-related nucleic acid drugs have demonstrated considerable potential in AD treatment; however, their clinical translation is limited by complex synthesis processes and carrier toxicity. Herein, an intranasally administrated, reactive oxygen species (ROS)-responsive, carrier-free gene delivery nanosystem (FTBR-NAC) was constructed for re-polarizing microglia and decreasing amyloid-β expression. In this nanosystem, fingolimod was conjugated with biguanide via an ROS-responsive linker to form the carrier for β-secretase 1 siRNA (siBACE1) to form FTBR nanoparticles. The electropositivity of FTBR and mucolytic activity of N-acetylcysteine (NAC) together enhanced the brain entry of FTBR. Upon reaching the brain, FTBR responded to the elevated ROS at the pathological site, releasing siBACE1 and fingolimod. Administration of FTBR-NAC improved cognitive function in AD mice, demonstrating the high therapeutic efficacy of this relatively simple nanosystem.
Collapse
Affiliation(s)
- Yongke Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610064, Sichuan, PR China
| | - Xiaotong Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610064, Sichuan, PR China
| | - Jie Li
- Department of Radiology and Huaxi MR Research Center, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610064, Sichuan, PR China
| | - Hang Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610064, Sichuan, PR China
| | - Qianqian Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610064, Sichuan, PR China
| | - Wenqing Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610064, Sichuan, PR China
| | - Ting Lei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610064, Sichuan, PR China
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610064, Sichuan, PR China
| | - Qiyong Gong
- Department of Radiology and Huaxi MR Research Center, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610064, Sichuan, PR China; Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361000, Fujian, PR China
| | - Hanmei Li
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Haoxing Wu
- Department of Radiology and Huaxi MR Research Center, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610064, Sichuan, PR China.
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610064, Sichuan, PR China.
| |
Collapse
|
40
|
McCrea LT, Batorsky RE, Bowen JJ, Yeh H, Thanos JM, Fu T, Perlis RH, Sheridan SD. Identifying brain-penetrant small-molecule modulators of human microglia using a cellular model of synaptic pruning. Neuropsychopharmacology 2025:10.1038/s41386-025-02123-1. [PMID: 40346178 DOI: 10.1038/s41386-025-02123-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/03/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025]
Abstract
Microglia dysregulation is implicated across a range of neurodevelopmental and neurodegenerative disorders, making their modulation a promising therapeutic target. Using PBMC-derived induced microglia-like cells (piMGLCs) in a scalable assay, we screened 489 CNS-penetrant compounds for modulation of microglial phagocytosis of human synaptosomes in a validated assay for microglia-mediated synaptic pruning. Compounds from the library that reduced phagocytosis by ≥2 standard deviations across the library without cytotoxicity were validated in secondary screens, with 28 of them further confirmed to reduce phagocytosis by 50% or more. These compounds comprise a wide range of therapeutic classes with different mechanisms of action, including immunosuppressants, kinase inhibitors, antipsychotics, and epigenetic modulators. Image-based morphological measurements were calculated to measure the degree of ramified vs. ameboid morphotypes as an indicator of activation state. Additionally, transcriptomic profiling indicated divergent effects on cell signaling, metabolism, activation, and actin dynamics across confirmed compounds. In particular, multiple CNS-penetrant small molecules with prior FDA approval or demonstration of safety in vivo demonstrated modulatory effects on microglia. For example, identified drugs such as the tyrosine kinase inhibitors lapatinib, alectinib, and lazertinib and the epigenetic modulator vorinostat have been approved for various cancer treatments and are being investigated for other indications; however, they have not been extensively studied in patients for neurodevelopmental and neurodegenerative disorders. These potential disease-modifying agents represent high-priority candidates for repositioning studies in neurodevelopmental, neuroinflammatory, or neurodegenerative disorders.
Collapse
Affiliation(s)
- Liam T McCrea
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca E Batorsky
- Tufts Institute for Artificial Intelligence, Tufts University, Medford, MA, USA
| | - Joshua J Bowen
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Hana Yeh
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Jessica M Thanos
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Ting Fu
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Roy H Perlis
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| | - Steven D Sheridan
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Bernier V, Chatelan A, Point C, Strauss M. Nutrition and Neuroinflammation: Are Middle-Aged Women in the Red Zone? Nutrients 2025; 17:1607. [PMID: 40431348 PMCID: PMC12113692 DOI: 10.3390/nu17101607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
Women exhibit unique vulnerabilities in health, especially regarding mental health and neurodegenerative diseases. Biological, hormonal, and metabolic differences contribute to sex-specific risks that remain underrepresented in clinical studies. Diseases such as major depressive disorder (MDD) and Alzheimer's disease (AD) are more prevalent in women and may be influenced by hormonal transitions, particularly during menopause. Chronic low-grade inflammation is emerging as a shared mechanism underlying both conditions, and this inflammatory state can be worsened by dietary habits. During menopause, mood and sleep disturbances can influence dietary behavior, leading to enhanced snacking and consumption of high-glycemic and comfort foods. Such foods, low in nutritional value, promote weight gain and elevated inflammatory markers. Their consumption combined (or not) with a preexisting Western diet pattern-already linked to inflammation-could reinforce systemic inflammation involving the gut-brain axis. Moreover, the symptoms "per se" could act on inflammation as well. Peripheral inflammation may cross the blood-brain barrier, sustaining mood disorders and promoting neurodegenerative changes. Finally, MDD and AD are both associated with conditions such as obesity and diabetes, which occur more frequently in women. The review highlights how menopause-related changes in mood, sleep, and diet may heighten susceptibility to mental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Veronique Bernier
- Department of Psychiatry, Brugmann University Hospital, Université Libre de Bruxelles—ULB, 1020 Brussels, Belgium
| | - Angeline Chatelan
- Department of Nutrition and Dietetics, Geneva School of Health Sciences, HES-SO University of Applied Sciences and Arts Western Switzerland, CH-1227 Geneva, Switzerland
| | - Camille Point
- Department of Psychiatry, Brugmann University Hospital, Université Libre de Bruxelles—ULB, 1020 Brussels, Belgium
| | - Mélanie Strauss
- Department of Neurology and Sleep Unit, Université Libre de Bruxelles—ULB, Hôpital Universitaire de Bruxelles (H.U.B), CUB Hôpital Erasme, Route de Lennik 808, 1070 Bruxelles, Belgium
- Laboratory of Experimental Neurology, Université Libre de Bruxelles—ULB, Route de Lennik 808, 1070 Bruxelles, Belgium
| |
Collapse
|
42
|
Du B, Zou Q, Wang X, Wang H, Yang X, Wang Q, Wang K. Multi-targeted engineered hybrid exosomes as Aβ nanoscavengers and inflammatory modulators for multi-pathway intervention in Alzheimer's disease. Biomaterials 2025; 322:123403. [PMID: 40347851 DOI: 10.1016/j.biomaterials.2025.123403] [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: 01/16/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
The pathogenesis of Alzheimer's disease (AD) was complex, including excessive deposition of β-amyloid (Aβ), microglia dysfunction, and neuroinflammation. Therefore, single-pathway treatment was not sufficient to ameliorate the multifaceted pathological changes associated with AD. Moreover, the low permeability of blood-brain barrier (BBB) and the lack of AD locus selectivity further limited the intervention efficacy of current AD drugs. In this study, a novel nanoparticle coating was designed by hybridizing the membrane from brain microvascular endothelial cell exosomes and macrophage exosomes, and combining polydopamine nanoparticles, resveratrol and Aβ-targeting aptamers to construct engineered exosomes (RPDA@Rb-A) with multiple targeting capabilities to intervene in Aβ clearance and regulate microglial dysfunction. Based on the homing effect of brain microvascular endothelial cell exosomes and the natural inflammation targeting ability of macrophage exosomes, RPDA@Rb-A can easily penetrate the blood brain barrier and accumulate in the brain inflammation site after capturing Aβ aggregates. RPDA@Rb-A can effectively intervene in AD through multi-pathway, including degraded toxic Aβ aggregates through local heating induced by near-infrared laser irradiation and alleviated neurotoxicity, promoted microglial clearance of Aβ by capturing Aβ, and modulated microglia-induced neuroinflammation by efficient delivery of small molecule drugs. In AD mouse model, the administration of RPDA@Rb-A resulted in a significant reduction in amyloid plaque deposition, neuroinflammation, and cognitive impairments. The engineered exosomes based on membrane hybridization overcome the shortcomings of traditional drug carriers in poor penetration and insufficient targeting to the central nervous system, and provide a potential platform for multi pathways intervention in AD.
Collapse
Affiliation(s)
- Bin Du
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, PR China
| | - Qingqing Zou
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, PR China
| | - Xin Wang
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, PR China
| | - Hongqiang Wang
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, PR China
| | - Xiaohai Yang
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, PR China
| | - Qing Wang
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, PR China.
| | - Kemin Wang
- State Key Laboratory of Chemo and Biosensing, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, 410082, PR China
| |
Collapse
|
43
|
Qin Q, Xia X, Qu J, Guan Z, Yin Y, Chang J, Yu C, Zhang T, Tang Y. Blood biomarkers of amyloid and tau pathologies, brain degeneration, inflammation, and oxidative stress in early- and late-onset Alzheimer's disease. J Alzheimers Dis 2025:13872877251340955. [PMID: 40336292 DOI: 10.1177/13872877251340955] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
BackgroundNumerous blood biomarkers have emerged as promising biomarkers for Alzheimer's disease (AD) and cognitive decline, but limited knowledge exists concerning the difference of blood biomarkers between early-onset and late-onset cases.ObjectiveInvestigate blood biomarkers associated with amyloid and tau pathologies, brain degeneration, inflammation, and oxidative stress in individuals afflicted with both early-onset and late-onset AD, as well as in age-matched healthy controls.MethodsA total of 125 participants were enrolled. We assessed levels of 18 distinct blood biomarkers and their associations with cerebrospinal fluid biomarkers, neuropsychological test scores, APOE ε4 carrier status, and neuroimaging markers. The diagnostic potential of blood biomarkers was investigated.ResultsIn early-onset AD patients, levels of blood Interleukin (IL)-4, IL-6, and Tumor necrosis factor-alpha (TNF-α) were notably lower comparing to late-onset patients. AD patients exhibited higher blood levels of phosphorylated-tau181 (p-tau181), neurofilament light chain (NfL), and glial fibrillary acidic protein (GFAP), as well as lower levels of amyloid-β (Aβ)42 and IL-12p70. Oxidative stress markers, including malondialdehyde, total antioxidant capacity, and superoxide dismutase, exhibited a progressive trend across the continuum of AD. Inflammatory markers demonstrating correlations with neuroimaging markers. Blood levels of Aβ42, p-tau181, NfL, and GFAP associated with neuropsychological scores and effectively discriminated AD, with GFAP exhibiting particular relevance in early-onset cases.ConclusionsInflammatory markers exhibited differences between patients with early- and late-onset AD, associated with alterations in brain structure and function. With the progression of disease continuum, a decrement in antioxidant capacity was observed. Blood Aβ42, p-tau181, NfL, and GFAP showed promise in detecting cognitive decline and AD.
Collapse
Affiliation(s)
- Qi Qin
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xinyi Xia
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Junda Qu
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Zhongtian Guan
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Yunsi Yin
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Chang
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chaoji Yu
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tongtong Zhang
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yi Tang
- Department of Neurology, Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
- Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing, China
| |
Collapse
|
44
|
Ali M, Garcia P, Lunkes LP, Sciortino A, Thomas MH, Heurtaux T, Grzyb K, Halder R, Skupin A, Buée L, Blum D, Buttini M, Glaab E. Temporal transcriptomic changes in the THY-Tau22 mouse model of tauopathy display cell type- and sex-specific differences. Acta Neuropathol Commun 2025; 13:93. [PMID: 40336141 PMCID: PMC12060421 DOI: 10.1186/s40478-025-02013-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Tauopathies, including Alzheimer's disease (AD) and frontotemporal dementia (FTD), display sex-specific differences in prevalence and progression, but the underlying molecular mechanisms remain unclear. Single-cell transcriptomic analysis of animal models can reveal how AD pathology affects different cell types across sex and age. OBJECTIVE To understand sex-specific and sex-dimorphic transcriptomic changes in different cell types and their age-dependence in the THY-Tau22 mouse model of AD-linked tauopathy. METHODS We applied single-cell RNA sequencing (scRNA-seq) to cortical tissue from male and female THY-Tau22 and wild-type mice at 17 months of age, when they had prominent tau inclusion pathology, and compared the results with corresponding data previously obtained at 7 months of age. Using differential statistical analysis for individual genes, pathways, and gene regulatory networks, we identified sex-specific, sex-dimorphic, and sex-neutral changes, and looked at how they evolved over age. To validate the most robust findings across distinct mouse models and species, the results were compared with cortical scRNA-seq data from the transgenic hAPP-based Tg2576 mouse model and human AD. RESULTS We identified several significant sex-specific and sex-dimorphic differentially expressed genes in neurons, microglia, astrocytes and oligodendrocytes, including both cross-sectional changes and alterations from 7 months to 17 months of age. Key pathways affected in a sex-dependent manner across age included neurotransmitter signaling, RNA processing and splicing, stress response pathways, and protein degradation pathways. In addition, network analysis revealed the AD-associated genes Clu, Mbp, Fos and Junb as relevant regulatory hubs. Analysis of age-dependent changes highlighted genes and pathways associated with inflammatory response (Malat1, Cx3cr1), protein homeostasis (Cst3), and myelin maintenance (Plp1, Cldn11, Mal) that showed consistent sex-dependent changes as the THY-Tau22 mice aged. Multiple genes with established implications in AD, including the long non-coding RNA gene Malat1, displayed concordant sex-specific changes in mouse models and human AD. CONCLUSIONS This study provides a comprehensive single-cell transcriptomic characterization of sex-linked and age-dependent changes in the THY-Tau22 tauopathy model, revealing new insights into the interplay between age-dependent AD-like pathologies and sex. The identified sex-specific changes and their conservation across models and human AD highlight molecular targets for further preclinical investigation of sex-specific therapeutic strategies in AD.
Collapse
Affiliation(s)
- Muhammad Ali
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Pierre Garcia
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Laetitia P Lunkes
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Alessia Sciortino
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Melanie H Thomas
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Tony Heurtaux
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4367, Luxembourg
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Luc Buée
- Lille Neuroscience & Cognition, University of Lille, Inserm, CHU Lille, Alzheimer & Tauopathies, LabEx DISTALZ, Lille, UMR-S1172, France
| | - David Blum
- Lille Neuroscience & Cognition, University of Lille, Inserm, CHU Lille, Alzheimer & Tauopathies, LabEx DISTALZ, Lille, UMR-S1172, France
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, L-4362, Luxembourg.
| |
Collapse
|
45
|
Ortiz AA, Murtishaw AS, Beckholt M, Salazar AM, Osse AML, Kinney JW. Impact of chronic hyperglycemia and high-fat diet on Alzheimer's disease-related pathology in CX3CR1 knockout mice. Metab Brain Dis 2025; 40:197. [PMID: 40332622 DOI: 10.1007/s11011-025-01618-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 04/18/2025] [Indexed: 05/08/2025]
Abstract
Diabetes mellitus (DM), obesity, and metabolic syndrome are related disorders with wide-ranging and devastating effects that are comorbid with many other diseases. Clinical and epidemiological studies have found that type II diabetes mellitus (T2DM), including chronic hyperglycemia and hyperinsulinemia, significantly increases the risk of Alzheimer's disease (AD) and other forms of dementia in the elderly. Insulin has slightly different functions in the peripheral body than in the central nervous system and the dysregulation of these functions may contribute to the onset and progression of late-life neurodegenerative disease. To investigate cognitive function and AD-related disease pathology, we utilized two different models of key features of diabetes, one model characterized by hyperglycemia resulting from a diabetogenic compound that selectively targets insulin-producing pancreatic β-cells, and the other model based on diet-induced obesity. Additionally, these diabetic models were combined with fractalkine receptor knockout mice (CX3CR1-/-), a genetic mouse model of inflammation, to explore the additive effects of multiple AD risk factors. The CX3CR1 receptor has been implicated in modulating neuroinflammation associated with AD, and its dysregulation can exacerbate metabolic disturbances and neurodegenerative markers. We found that diabetic-status, regardless of whether it was drug- or diet-induced, resulted in profound impairments in learning and memory and AD-related alterations within the hippocampus.
Collapse
Affiliation(s)
- Andrew Adonay Ortiz
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA.
| | - Andrew Scott Murtishaw
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Monica Beckholt
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Arnold Maloles Salazar
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Amanda Marie Leisgang Osse
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA
| | - Jefferson William Kinney
- Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Las Vegas, NV, 89154, USA
| |
Collapse
|
46
|
Auger A, Faidi R, Rickman AD, Martinez CP, Fajfer A, Carling J, Hilyard A, Ali M, Ono R, Cleveland C, Seliniotakis R, Truong N, Chefson A, Raymond M, Germain MA, Crackower MA, Heckmann BL. Post-symptomatic NLRP3 inhibition rescues cognitive impairment and mitigates amyloid and tau driven neurodegeneration. NPJ DEMENTIA 2025; 1:3. [PMID: 40343261 PMCID: PMC12055592 DOI: 10.1038/s44400-025-00011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
Emerging evidence has established neuroinflammation as a primary driver of progressive neuronal loss observed across neurodegenerative diseases (NDDs). The NLRP3 inflammasome is a cytosolic immunoprotective danger sensing complex, which when aberrantly activated drives neuroinflammation, propagates amyloid deposition, and neurodegeneration. Herein, we report the therapeutic benefit of NLRP3 inflammasome inhibition in Alzheimer's disease (AD), using a novel and selective brain-penetrant small molecule NLRP3 inhibitor, VEN-02XX, which we profiled in the 5XFAD/Rubicon KO AD model. We demonstrate for the first time that targeting NLRP3, post-symptomatic establishment, rescues cognitive deficits, mitigates neuronal loss, and is sufficient to significantly reduce reactive microgliosis, neuroinflammation and tau pathology. Our data further suggest that pharmacological inhibition of NLRP3, after disease onset, has the potential to reduce cortical and hippocampal amyloid burden. Together, these results highlight the potential for NLRP3 inhibition as a symptomatic and disease modifying therapeutic target for AD pathology and more broadly NDDs.
Collapse
Affiliation(s)
- Anick Auger
- Ventus Therapeutics, Inc., 4800 Rue Levy, Montreal, QC, H4R 2P7 Canada
| | - Rania Faidi
- Ventus Therapeutics, Inc., 4800 Rue Levy, Montreal, QC, H4R 2P7 Canada
| | - Alexis D. Rickman
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| | - Carolina Pena Martinez
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| | - Austin Fajfer
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| | - Jeremy Carling
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| | - Addison Hilyard
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| | - Mubashshir Ali
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| | - Ryosuke Ono
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| | - Connor Cleveland
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| | - Ria Seliniotakis
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| | - Nhi Truong
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| | - Amandine Chefson
- Ventus Therapeutics, Inc., 4800 Rue Levy, Montreal, QC, H4R 2P7 Canada
| | - Marianne Raymond
- Ventus Therapeutics, Inc., 4800 Rue Levy, Montreal, QC, H4R 2P7 Canada
| | | | - Michael A. Crackower
- Ventus Therapeutics U.S., Inc., 100 Beaver Street, Suite 201, Waltham, MA 02453 USA
| | - Bradlee L. Heckmann
- USF Health Byrd Alzheimer’s Center and Neuroscience Institute, Department of Molecular Medicine Morsani College of Medicine, Tampa, FL 33613 USA
| |
Collapse
|
47
|
Berrocal M, Alvarez-Barrientos A, Mata AM. Neurotoxic amyloid β-peptide and tau produce cytokine-like effects on PMCA in glioblastoma cell lines, enhancing its activity and isoforms expression. FEBS Open Bio 2025. [PMID: 40325855 DOI: 10.1002/2211-5463.70046] [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: 01/14/2025] [Revised: 02/14/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025] Open
Abstract
The transformation of astrocytes into neurotoxic reactive astrocytes, classified as A1, by inflammatory cytokines, and their link to brain damage and neurodegenerative diseases has been widely documented. However, the roles of two biomarkers of Alzheimer's disease (AD), amyloid β-peptide (Aβ) and tau, and that of calcium pumps which are involved in the fine-tuning of calcium homeostasis, are poorly understood in astrocytes. In this study, we showed that treating astrocytoma U-251 cells with a cocktail of cytokines significantly increased plasma membrane Ca2+-ATPase (PMCA) activity and expression levels of the four PMCA isoforms. Moreover, treatment of cells with Aβ1-42 or tau induced a similar upregulation of PMCA activity and isoform expression levels as cytokines. These effects support the close association of Aβ and tau with inflammation. This study may help better understand the role of PMCA in promoting calcium extrusion from astrocytes transformed by AD markers.
Collapse
Affiliation(s)
- María Berrocal
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
- Instituto de Biomarcadores de Patologías Moleculares (IBPM), Universidad de Extremadura, Badajoz, Spain
| | - Alberto Alvarez-Barrientos
- Servicio de Técnicas Aplicadas a la Biociencia (STAB), Edificio Guadiana, SAIUEx, Universidad de Extremadura, Badajoz, Spain
| | - Ana M Mata
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
- Instituto de Biomarcadores de Patologías Moleculares (IBPM), Universidad de Extremadura, Badajoz, Spain
| |
Collapse
|
48
|
Borda MG, Ramírez-Vélez R, Botero-Rodriguez F, Patricio-Baldera J, de Lucia C, Pola I, Barreto GE, Khalifa K, Bergland AK, Kivipelto M, Cederholm T, Zetterberg H, Ashton NJ, Ballard C, Siow R, Aarsland D. Anthocyanin supplementation in adults at risk for dementia: a randomized controlled trial on its cardiometabolic and anti-inflammatory biomarker effects. GeroScience 2025:10.1007/s11357-025-01669-8. [PMID: 40314845 DOI: 10.1007/s11357-025-01669-8] [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: 02/20/2025] [Accepted: 04/15/2025] [Indexed: 05/03/2025] Open
Abstract
Anthocyanins are dietary flavonoids shown to have a therapeutic capacity to mitigate inflammation and oxidative stress. The present secondary analyses from the "Anthocyanins in People at Risk for Dementia Study" were aimed at (I) determining the intervention's effect on blood-based markers of cardiovascular disease and inflammation and (II) evaluating whether baseline factors such as age, sex, inflammation, or cardiometabolic score may moderate the intervention's effect on inflammatory status. This study was an ancillary, 24-week randomized, double-blind, placebo-controlled Phase II trial. Sub-sample participants (n = 99), aged 60-80 years with mild cognitive impairment or cardiometabolic disorders, were randomized to receive either 320 mg/day of anthocyanins or placebo. The biomarkers analyzed included inflammatory biomarker assessment (IL - 6, IL - 8, IL - 10, IL - 1b, TNF - α, IFN - γ), and C-reactive protein (CRP), as well as albumin, thrombocytes, cholesterol, LDL, HDL, and triglycerides, which were longitudinally compared between both groups. Baseline characteristics were balanced between the groups. ANCOVA analyses reveal 24-week differences favoring the anthocyanin treatment in LDL cholesterol levels (ƞp2 = 0.078; p = 0.015), cardiometabolic score (ƞp2 = 0.073; p = 0.021), CRP levels (ƞp2 = 0.417; p = 0.0001), IL - 6 (ƞp2 = 0.085; p = 0.015), IL - 1b (ƞp2 = 0.058; p = 0.037), and Inflam z-score 5 (ƞp2 = 0.059, p = 0.004). Moderation analysis demonstrated that the inflammatory score at baseline was a significant predictor of the effect of the intervention on the CRP levels. Anthocyanin supplementation reduces CRP and cardiovascular disease biomarkers in individuals at risk of dementia, especially when there is increased inflammation at baseline. ClinicalTrials.gov study identifier: NCT03419039.
Collapse
Affiliation(s)
- Miguel German Borda
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway.
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain.
- Centro de Investigación en Ciencias de La Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan Edo. de México, México.
| | - Robinson Ramírez-Vélez
- Navarrabiomed, IdiSNA, Hospital Universitario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Felipe Botero-Rodriguez
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- SynaptIA - Inteligencia artificial para la investigación en salud mental, Bogotá, Colombia
- Centro de Memoria y Cognición Intellectus, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Jonathan Patricio-Baldera
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Instituto de Investigación en Salud, Facultad de Ciencias de La Salud de La Universidad Autónoma de Santo Domingo, Santo Domingo, Dominican Republic
| | - Chiara de Lucia
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Ilaria Pola
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Khadija Khalifa
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
| | - Anne Katrine Bergland
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Tommy Cederholm
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, 62167, Uppsala, Sweden
- Theme Inflammation & Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK, Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Nicholas J Ashton
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Banner Alzheimer's Institute, University of Arizona, Phoenix, AZ, USA
| | - Clive Ballard
- Medical School, University of Exeter, University of Exeter, Exeter, UK
| | - Richard Siow
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Dag Aarsland
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, Helse Stavanger HF, Postboks 8100, 4068, Stavanger, Norway
- Centre for Healthy Brain Ageing, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| |
Collapse
|
49
|
Mimmi S, Parrotta EI, Tolomeo AM, Maisano D, Crapella V, Pingitore E, Fatima K, Zimbo AM, Talarico M, Cristiani CM, Scaramuzzino L, Valente D, Zannino C, Cuda G, Quattrone A, Iaccino E, Quattrone A. Neuronally-Derived Extracellular Vesicles Transforming Growth Factor Beta-1 Levels in Progressive Supranuclear Palsy. Mov Disord 2025. [PMID: 40317792 DOI: 10.1002/mds.30222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/26/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Differentiating progressive supranuclear palsy (PSP) from other parkinsonian disorders may be challenging. OBJECTIVES To investigate the role of transforming growth factor beta-1 (TGFβ1) in PSP. METHODS A total of 33 PSP, 39 Parkinson's disease (PD), 8 multiple system atrophy (MSA) patients, and 50 healthy controls (HC) were enrolled. TGFβ1 levels, including both active and inactive forms (latency-associated peptide [LAP]-TGFβ1), were measured in serum, total extracellular vesicles (EVs), and neuronally-derived EVs (NDEVs) using microfluidic assays and ELISA. RESULTS PSP patients exhibited a marked increase in TGFβ1 and LAP-TGFβ1 levels in NDEVs, while no differences were observed across groups in serum or total EVs. Receiver operating characteristic (ROC) analysis demonstrated outstanding performance in differentiating PSP from non-PSP patients (TGFβ1, area under the curve [AUC]: 0.97; LAP-TGFβ1, AUC: 1.00), HC, AUC: 1.00). CONCLUSIONS This study highlights TGFβ1 and LAP-TGFβ1 in NDEVs as promising blood-based non-invasive biomarkers for PSP diagnosis, paving the way for further research on these proteins in PSP. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Selena Mimmi
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | | | - Anna Maria Tolomeo
- Department of Cardiac, Thoracic and Vascular Science and Public Health, University of Padova, Padua, Italy
- Institute of Pediatric Research 'Città della Speranza', Padua, Italy
| | | | - Valentina Crapella
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Elisabetta Pingitore
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Khushboo Fatima
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Anna Maria Zimbo
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Mariagrazia Talarico
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Costanza Maria Cristiani
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Luana Scaramuzzino
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Desirèe Valente
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Clara Zannino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Gianni Cuda
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Aldo Quattrone
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Enrico Iaccino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Andrea Quattrone
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
- Institute of Neurology, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
50
|
Vollhardt A, Frölich L, Stockbauer AC, Danek A, Schmitz C, Wahl AS. Towards a better diagnosis and treatment of dementia: Identifying common and distinct neuropathological mechanisms in Alzheimer's and vascular dementia. Neurobiol Dis 2025; 208:106845. [PMID: 39999928 DOI: 10.1016/j.nbd.2025.106845] [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: 11/08/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) together contribute to almost 90 % of all dementia cases leading to major health challenges of our time with a substantial global socioeconomic burden. While in AD, the improved understanding of Amyloid beta (Aß) mismetabolism and tau hyperphosphorylation as pathophysiological hallmarks has led to significant clinical breakthroughs, similar advances in VaD are lacking. After comparing the clinical presentation, including risk factors, disease patterns, course of diseases and further diagnostic parameters for both forms of dementia, we highlight the importance of shared pathomechanisms found in AD and VaD: Endothelial damage, blood brain barrier (BBB) breakdown and hypoperfusion inducing oxidative stress and inflammation and thus trophic uncoupling in the neurovascular unit. A dysfunctional endothelium and BBB lead to the accumulation of neurotoxic molecules and Aß through impaired clearance, which in turn leads to neurodegeneration. In this context we discuss possible neuropathological parameters, which might serve as biomarkers and thus improve diagnostic accuracy or reveal targets for novel therapeutic strategies for both forms of dementia.
Collapse
Affiliation(s)
- Alisa Vollhardt
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Lutz Frölich
- Central Institute of Mental Health, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Anna Christina Stockbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Adrian Danek
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Christoph Schmitz
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Anna-Sophia Wahl
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany; Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany.
| |
Collapse
|