Other Sites:
Robert J. Robbins is a biologist, an educator, a science administrator, a publisher, an information technologist, and an IT leader and manager who specializes in advancing biomedical knowledge and supporting education through the application of information technology. More About: RJR | OUR TEAM | OUR SERVICES | THIS WEBSITE
RJR: Recommended Bibliography 29 Nov 2025 at 01:36 Created:
Alzheimer Disease — Current Literature
Alzheimer's disease is an irreversible, progressive brain disorder that slowly destroys memory and thinking skills, and eventually the ability to carry out the simplest tasks. In most people with Alzheimer's, symptoms first appear in their mid-60s. Alzheimer's is the most common cause of dementia among older adults. Dementia is the loss of cognitive functioning — thinking, remembering, and reasoning — and behavioral abilities to such an extent that it interferes with a person's daily life and activities. Dementia ranges in severity from the mildest stage, when it is just beginning to affect a person's functioning, to the most severe stage, when the person must depend completely on others for basic activities of daily living. Scientists don't yet fully understand what causes Alzheimer's disease in most people. There is a genetic component to some cases of early-onset Alzheimer's disease. Late-onset Alzheimer's arises from a complex series of brain changes that occur over decades. The causes probably include a combination of genetic, environmental, and lifestyle factors. The importance of any one of these factors in increasing or decreasing the risk of developing Alzheimer's may differ from person to person. This bibliography runs a generic query on "Alzheimer" and then restricts the results to papers published in or after 2017.
Created with PubMed® Query: 2023:2025[dp] AND ( alzheimer*[TIAB] ) NOT pmcbook NOT ispreviousversion
Citations The Papers (from PubMed®)
RevDate: 2025-11-28
Nanocarrier-Enhanced Simvastatin Modulates AMPK-ULK1 Pathway and Oxidative Stress in Alzheimer's Disease Model.
European journal of pharmacology pii:S0014-2999(25)01165-3 [Epub ahead of print].
In Alzheimer's disease (AD), the hippocampus and cerebral cortex are primarily affected, showing degeneration of cholinergic neurons and Aβ plaques accumulation. These changes are strongly associated with oxidative stress, neuroinflammation, and impaired autophagy. In our study simvastatin (Simva), a lipid-lowering statin, was investigated against aluminum chloride (AlCl3)-induced rat model of AD and showed potential neuroprotective effects. Nano-delivery systems were used to enhance Simva's brain pharmacological activity. Adult male Sprague Dawley rats were designated into four groups: Control group (normal saline, IP, 28 days), AlCl3 group (25 mg/ kg AlCl3, IP for 28 days), Simva group (Simva 10 mg/kg, orally for 28 days 1 hr before AlCl3 injection) and Simva-Nano group (Simva nanocarrier 10mg/kg, orally for 28 days 1 hr before AlCl3 injection). Simva and its nanocarrier enhanced autophagy by upregulating p-AMPK, p-ULK1, and LC3-II while downregulating ULK1. They showed strong antioxidant and anti-inflammatory effects by decreasing MDA, TNF-α, IL-1β and increased GSH. Additionally, they increased ACh levels and suppressed AChE gene expression. Immunohistochemical staining revealed substantial reduction in Aβ plaques in the Simva group, while in Simva-Nano group, Aβ plaques were not detected under our assay conditions. Aβ immunostaining was quantified as the number of Aβ deposits per field (6 fields). Histopathological analysis confirmed reduced neuronal degeneration and glial activation, with the Simva-Nano group demonstrating near-normal hippocampal architecture. In conclusion, Simva exhibits notable neuroprotective effects in the AD rat model via antioxidative, anti-inflammatory, and autophagic pathways. Nanocarrier delivery further improves Simva's pharmacological effects, offering a promising AD therapeutic strategy.
Additional Links: PMID-41314452
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41314452,
year = {2025},
author = {Afify, AM and El-Sayed, EK and Ismail, A and Nofal, S},
title = {Nanocarrier-Enhanced Simvastatin Modulates AMPK-ULK1 Pathway and Oxidative Stress in Alzheimer's Disease Model.},
journal = {European journal of pharmacology},
volume = {},
number = {},
pages = {178411},
doi = {10.1016/j.ejphar.2025.178411},
pmid = {41314452},
issn = {1879-0712},
abstract = {In Alzheimer's disease (AD), the hippocampus and cerebral cortex are primarily affected, showing degeneration of cholinergic neurons and Aβ plaques accumulation. These changes are strongly associated with oxidative stress, neuroinflammation, and impaired autophagy. In our study simvastatin (Simva), a lipid-lowering statin, was investigated against aluminum chloride (AlCl3)-induced rat model of AD and showed potential neuroprotective effects. Nano-delivery systems were used to enhance Simva's brain pharmacological activity. Adult male Sprague Dawley rats were designated into four groups: Control group (normal saline, IP, 28 days), AlCl3 group (25 mg/ kg AlCl3, IP for 28 days), Simva group (Simva 10 mg/kg, orally for 28 days 1 hr before AlCl3 injection) and Simva-Nano group (Simva nanocarrier 10mg/kg, orally for 28 days 1 hr before AlCl3 injection). Simva and its nanocarrier enhanced autophagy by upregulating p-AMPK, p-ULK1, and LC3-II while downregulating ULK1. They showed strong antioxidant and anti-inflammatory effects by decreasing MDA, TNF-α, IL-1β and increased GSH. Additionally, they increased ACh levels and suppressed AChE gene expression. Immunohistochemical staining revealed substantial reduction in Aβ plaques in the Simva group, while in Simva-Nano group, Aβ plaques were not detected under our assay conditions. Aβ immunostaining was quantified as the number of Aβ deposits per field (6 fields). Histopathological analysis confirmed reduced neuronal degeneration and glial activation, with the Simva-Nano group demonstrating near-normal hippocampal architecture. In conclusion, Simva exhibits notable neuroprotective effects in the AD rat model via antioxidative, anti-inflammatory, and autophagic pathways. Nanocarrier delivery further improves Simva's pharmacological effects, offering a promising AD therapeutic strategy.},
}
RevDate: 2025-11-28
Review of Artificial Intelligence for Clinical Use in Alzheimer's Disease and Related Dementias.
Seminars in neurology [Epub ahead of print].
As the U.S. population ages, Alzheimer's disease and related dementias (ADRD) cases are increasing, resulting in long wait times for specialist care. We review state-of-the-art artificial intelligence (AI) applications in ADRD care, from streamlining clinical diagnosis to pioneering novel digital biomarkers. Near-term AI applications include neuroimaging interpretation, conversational agents for patient interviews, and digital cognitive assessments. Large language models show promise as collaborative partners, helping clinicians interpret complex data while supporting patients and caregivers. Emerging digital biomarkers-speech analysis, passive monitoring through wearable devices, electronic health record analysis, and multiomics-offer potential for continuous monitoring to detect cognitive decline years before traditional assessments. Despite the acceleration of AI innovation, most of these systems are inaccessible in clinical practice. Implementation bottlenecks include limited external validation, technical challenges, model biases, infrastructure, and regulatory requirements. This review aims to help neurologists navigate this rapidly evolving AI landscape and prepare for implementation in ADRD care.
Additional Links: PMID-41314410
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41314410,
year = {2025},
author = {Breithaupt, AG and Tang, A and Paolillo, EW and Bibars, M and Johnson, ECB and Saloner, R and Possin, KL and Windon, CC and Hill-Jarrett, TG and Giorgio, J and Rauschecker, AM and Kwon, H and Vonk, JMJ and Pinheiro-Chagas, P},
title = {Review of Artificial Intelligence for Clinical Use in Alzheimer's Disease and Related Dementias.},
journal = {Seminars in neurology},
volume = {},
number = {},
pages = {},
doi = {10.1055/a-2744-9871},
pmid = {41314410},
issn = {1098-9021},
support = {K23AG093166//National Institute on Aging of the National Institutes of Health/ ; },
abstract = {As the U.S. population ages, Alzheimer's disease and related dementias (ADRD) cases are increasing, resulting in long wait times for specialist care. We review state-of-the-art artificial intelligence (AI) applications in ADRD care, from streamlining clinical diagnosis to pioneering novel digital biomarkers. Near-term AI applications include neuroimaging interpretation, conversational agents for patient interviews, and digital cognitive assessments. Large language models show promise as collaborative partners, helping clinicians interpret complex data while supporting patients and caregivers. Emerging digital biomarkers-speech analysis, passive monitoring through wearable devices, electronic health record analysis, and multiomics-offer potential for continuous monitoring to detect cognitive decline years before traditional assessments. Despite the acceleration of AI innovation, most of these systems are inaccessible in clinical practice. Implementation bottlenecks include limited external validation, technical challenges, model biases, infrastructure, and regulatory requirements. This review aims to help neurologists navigate this rapidly evolving AI landscape and prepare for implementation in ADRD care.},
}
RevDate: 2025-11-28
Deep learning-derived arterial input function for dynamic brain PET.
NeuroImage pii:S1053-8119(25)00612-3 [Epub ahead of print].
Dynamic positron emission tomography (PET) imaging combined with radiotracer kinetic modeling is a powerful technique for visualizing biological processes in the brain, offering valuable insights into brain functions and neurological disorders such as Alzheimer's and Parkinson's diseases. Accurate kinetic modeling relies heavily on the use of a metabolite-corrected arterial input function (AIF), which typically requires invasive and labor-intensive arterial blood sampling. While alternative non-invasive approaches have been proposed, they often compromise accuracy or still necessitate at least one invasive blood sampling. In this study, we present the deep learning-derived arterial input function (DLIF), a deep learning framework capable of estimating a metabolite-corrected AIF directly from dynamic PET image sequences without any blood sampling. We validated DLIF using existing dynamic PET patient data. We compared DLIF and resulting parametric maps against ground truth measurements. Our evaluation shows that DLIF achieves accurate and robust AIF estimation. By leveraging deep learning's ability to capture complex temporal dynamics and incorporating prior knowledge of typical AIF shapes through basis functions, DLIF provides a rapid, accurate, and entirely non-invasive alternative to traditional AIF measurement methods.
Additional Links: PMID-41314271
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41314271,
year = {2025},
author = {Chen, J and Jiang, Z and Coughlin, JM and Cheong, I and Mills, KA and Pomper, MG and Du, Y},
title = {Deep learning-derived arterial input function for dynamic brain PET.},
journal = {NeuroImage},
volume = {},
number = {},
pages = {121609},
doi = {10.1016/j.neuroimage.2025.121609},
pmid = {41314271},
issn = {1095-9572},
abstract = {Dynamic positron emission tomography (PET) imaging combined with radiotracer kinetic modeling is a powerful technique for visualizing biological processes in the brain, offering valuable insights into brain functions and neurological disorders such as Alzheimer's and Parkinson's diseases. Accurate kinetic modeling relies heavily on the use of a metabolite-corrected arterial input function (AIF), which typically requires invasive and labor-intensive arterial blood sampling. While alternative non-invasive approaches have been proposed, they often compromise accuracy or still necessitate at least one invasive blood sampling. In this study, we present the deep learning-derived arterial input function (DLIF), a deep learning framework capable of estimating a metabolite-corrected AIF directly from dynamic PET image sequences without any blood sampling. We validated DLIF using existing dynamic PET patient data. We compared DLIF and resulting parametric maps against ground truth measurements. Our evaluation shows that DLIF achieves accurate and robust AIF estimation. By leveraging deep learning's ability to capture complex temporal dynamics and incorporating prior knowledge of typical AIF shapes through basis functions, DLIF provides a rapid, accurate, and entirely non-invasive alternative to traditional AIF measurement methods.},
}
RevDate: 2025-11-28
Rocket-inspired gas-propelled microneedles engineered with borneol-NLCs-loaded hierarchical cavities for enhanced brain delivery in Alzheimer's therapy.
International journal of pharmaceutics pii:S0378-5173(25)01255-4 [Epub ahead of print].
Microneedle technology has emerged as a promising transdermal platform for transdermal delivery of Alzheimer's disease therapeutics. However, conventional passive diffusion approaches face fundamental limitations in overcoming both skin and blood-brain barriers. To address these limitations, we developed a rocket-inspired gas-propelled microneedle system integrating nanostructured lipid carriers for enhanced dual-barrier penetration. The study combined fluid vortex generation with blood-brain barrier endothelial remodeling to achieve efficient drug delivery. Specifically, the system featured a hierarchical cavity design with borneol-modified huperzine A-loaded nanostructured lipid carriers (NLCs) (particle size: 89.6 ± 0.7 nm; zeta potential: -22.5 ± 0.5 mV; Hup A encapsulation efficiency: 83.40 ± 1.51 %; drug loading capacity: 2.63 ± 0.06 %) in primary cavities and a spatially isolated pneumatic initiator (ascorbic acid/sodium bicarbonate) in secondary cavities. Through multiphysics simulation coupled fluid dynamics with chemical reaction kinetics and mass transport phenomena, which revealed that acid-base reactions generate CO2 microbubbles capable of producing rapid thrust forces. This microbubble-mediated propulsion mechanism achieved a 50 % increase in penetration depth (up to ∼ 1428 µm) relative to conventional microneedle platforms. Remarkably, this innovative hierarchical cavity design achieved a high drug loading capacity of 182 μg/array while preserving spatial isolation between the reactants and therapeutic payload. In vivo studies validated efficient brain delivery, as evidenced by a marked increase (p < 0.001) in cortical acetylcholine concentrations and amelioration of scopolamine-induced spatial memory impairments in rat models. This work established a preclinical proof-of-concept for the enhanced brain delivery of therapeutics in neurodegenerative disease through the synergistic integration of gas-propulsion physics and advanced nanocarrier engineering, offering new possibilities for overcoming biological delivery barriers.
Additional Links: PMID-41314266
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41314266,
year = {2025},
author = {Shen, S and Zheng, Y and Xie, Y and You, X and Chen, X and Xie, B and Weng, J and Huo, Q and Zhang, Y and Meselhy Zayed, MR and Yan, Q},
title = {Rocket-inspired gas-propelled microneedles engineered with borneol-NLCs-loaded hierarchical cavities for enhanced brain delivery in Alzheimer's therapy.},
journal = {International journal of pharmaceutics},
volume = {},
number = {},
pages = {126418},
doi = {10.1016/j.ijpharm.2025.126418},
pmid = {41314266},
issn = {1873-3476},
abstract = {Microneedle technology has emerged as a promising transdermal platform for transdermal delivery of Alzheimer's disease therapeutics. However, conventional passive diffusion approaches face fundamental limitations in overcoming both skin and blood-brain barriers. To address these limitations, we developed a rocket-inspired gas-propelled microneedle system integrating nanostructured lipid carriers for enhanced dual-barrier penetration. The study combined fluid vortex generation with blood-brain barrier endothelial remodeling to achieve efficient drug delivery. Specifically, the system featured a hierarchical cavity design with borneol-modified huperzine A-loaded nanostructured lipid carriers (NLCs) (particle size: 89.6 ± 0.7 nm; zeta potential: -22.5 ± 0.5 mV; Hup A encapsulation efficiency: 83.40 ± 1.51 %; drug loading capacity: 2.63 ± 0.06 %) in primary cavities and a spatially isolated pneumatic initiator (ascorbic acid/sodium bicarbonate) in secondary cavities. Through multiphysics simulation coupled fluid dynamics with chemical reaction kinetics and mass transport phenomena, which revealed that acid-base reactions generate CO2 microbubbles capable of producing rapid thrust forces. This microbubble-mediated propulsion mechanism achieved a 50 % increase in penetration depth (up to ∼ 1428 µm) relative to conventional microneedle platforms. Remarkably, this innovative hierarchical cavity design achieved a high drug loading capacity of 182 μg/array while preserving spatial isolation between the reactants and therapeutic payload. In vivo studies validated efficient brain delivery, as evidenced by a marked increase (p < 0.001) in cortical acetylcholine concentrations and amelioration of scopolamine-induced spatial memory impairments in rat models. This work established a preclinical proof-of-concept for the enhanced brain delivery of therapeutics in neurodegenerative disease through the synergistic integration of gas-propulsion physics and advanced nanocarrier engineering, offering new possibilities for overcoming biological delivery barriers.},
}
RevDate: 2025-11-28
Neurodegenerative Disease and Autophagy in iPSC models.
Neuroscience research pii:S0168-0102(25)00174-9 [Epub ahead of print].
Neurodegenerative diseases are characterized by the gradual deterioration of specific neuronal populations, ultimately resulting in motor, cognitive, or behavioral impairments. Despite the worldwide increase in disease incidence, effective therapies remain unavailable. A common pathological hallmark of neurodegenerative diseases is the accumulation of misfolded protein aggregates, which impair normal cellular function. Accordingly, numerous studies and therapeutic strategies have focused on targeting these toxic aggregates and protein quality control via autophagy, a vital cellular recycling mechanism. Autophagy dysregulation has been implicated in the pathogenesis of several neurodegenerative diseases. Induced pluripotent stem cell (iPSC) technology has emerged as a powerful platform for modeling neurodegenerative diseases, and iPSC-derived models provide human-relevant systems for studying autophagic dysfunction in vitro. In this review, we discuss the key findings of recent studies investigating autophagy in iPSC-based models of neurodegenerative diseases, including Alzheimer's disease, amyotrophic lateral sclerosis, frontotemporal dementia, and other diseases.
Additional Links: PMID-41314255
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41314255,
year = {2025},
author = {Odonchimed, S and Imamura, K and Inoue, H},
title = {Neurodegenerative Disease and Autophagy in iPSC models.},
journal = {Neuroscience research},
volume = {},
number = {},
pages = {104991},
doi = {10.1016/j.neures.2025.104991},
pmid = {41314255},
issn = {1872-8111},
abstract = {Neurodegenerative diseases are characterized by the gradual deterioration of specific neuronal populations, ultimately resulting in motor, cognitive, or behavioral impairments. Despite the worldwide increase in disease incidence, effective therapies remain unavailable. A common pathological hallmark of neurodegenerative diseases is the accumulation of misfolded protein aggregates, which impair normal cellular function. Accordingly, numerous studies and therapeutic strategies have focused on targeting these toxic aggregates and protein quality control via autophagy, a vital cellular recycling mechanism. Autophagy dysregulation has been implicated in the pathogenesis of several neurodegenerative diseases. Induced pluripotent stem cell (iPSC) technology has emerged as a powerful platform for modeling neurodegenerative diseases, and iPSC-derived models provide human-relevant systems for studying autophagic dysfunction in vitro. In this review, we discuss the key findings of recent studies investigating autophagy in iPSC-based models of neurodegenerative diseases, including Alzheimer's disease, amyotrophic lateral sclerosis, frontotemporal dementia, and other diseases.},
}
RevDate: 2025-11-28
Therapeutic prospects of modulating TLR4/MAPK/ROS signalling in obesity-associated neuroinflammation.
Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 193:118805 pii:S0753-3322(25)00999-0 [Epub ahead of print].
It is becoming more widely acknowledged that obesity is a chronic low-grade inflammatory disease that has a significant influence on brain health in addition to metabolic problems. Adipose tissue growth, macrophage polarization, and cytokine release all contribute to systemic inflammation, which weakens the blood-brain barrier (BBB) and promotes immune-to-brain communication. Saturated fatty acids and gut-derived lipopolysaccharides activate Toll-like receptor 4 (TLR4) in the central nervous system, which triggers downstream nuclear factor-κB (NF-κB) and Mitogen-activated protein kinase (MAPK) cascades and increases neuroinflammation. At the same time, mitochondrial malfunction and oxidative stress hasten the buildup of reactive oxygen species (ROS), which further primes the NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome and maintains glial hyperactivation. These processes work together to cause synaptic dysfunction, insulin resistance in neurons, and heightened susceptibility to neurodegenerative illnesses, including Parkinson's and Alzheimer's. Pharmacological inhibitors, natural substances, and lifestyle changes that target TLR4, MAPK signaling, and ROS-mediated pathways have the potential to disrupt this metabolic-inflammatory-neuronal axis. Developing comprehensive solutions to reduce obesity-driven neuroinflammation requires an understanding of the molecular interactions between peripheral metabolic stress and central immune activation.
Additional Links: PMID-41314100
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41314100,
year = {2025},
author = {Sharma, D and Ravi, RN and Abdullah, ADI and Subramaniyan, V},
title = {Therapeutic prospects of modulating TLR4/MAPK/ROS signalling in obesity-associated neuroinflammation.},
journal = {Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie},
volume = {193},
number = {},
pages = {118805},
doi = {10.1016/j.biopha.2025.118805},
pmid = {41314100},
issn = {1950-6007},
abstract = {It is becoming more widely acknowledged that obesity is a chronic low-grade inflammatory disease that has a significant influence on brain health in addition to metabolic problems. Adipose tissue growth, macrophage polarization, and cytokine release all contribute to systemic inflammation, which weakens the blood-brain barrier (BBB) and promotes immune-to-brain communication. Saturated fatty acids and gut-derived lipopolysaccharides activate Toll-like receptor 4 (TLR4) in the central nervous system, which triggers downstream nuclear factor-κB (NF-κB) and Mitogen-activated protein kinase (MAPK) cascades and increases neuroinflammation. At the same time, mitochondrial malfunction and oxidative stress hasten the buildup of reactive oxygen species (ROS), which further primes the NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome and maintains glial hyperactivation. These processes work together to cause synaptic dysfunction, insulin resistance in neurons, and heightened susceptibility to neurodegenerative illnesses, including Parkinson's and Alzheimer's. Pharmacological inhibitors, natural substances, and lifestyle changes that target TLR4, MAPK signaling, and ROS-mediated pathways have the potential to disrupt this metabolic-inflammatory-neuronal axis. Developing comprehensive solutions to reduce obesity-driven neuroinflammation requires an understanding of the molecular interactions between peripheral metabolic stress and central immune activation.},
}
RevDate: 2025-11-28
CB1 receptor activation and inhibition differentially modulate cognitive deficits and neuropathology in 3xTg-AD mice.
Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 193:118818 pii:S0753-3322(25)01012-1 [Epub ahead of print].
Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) deposition, tau hyperphosphorylation, neuroinflammation, oxidative stress, and progressive neuronal loss. The endocannabinoid system regulates synaptic function, inflammation, and redox homeostasis through cannabinoid receptor type 1 (CB1). This study aimed to determine whether pharmacological activation or inhibition of the CB1 receptor differentially modulates Alzheimer's disease-related pathology. 3xTg-AD mice received weekly intraperitoneal injections of the CB1 agonist ACEA (1 mg/kg) or the inverse agonist AM251 (1 mg/kg) from 6 to 12 months of age. Cognitive function was assessed using the Morris Water Maze (MWM) and Y-maze, while hippocampal tissues were analyzed for Aβ, p-Tau, glial markers (GFAP, Iba-1), cytokines (IL-1β, IL-10), oxidative stress markers (SOD, GSH, MDA), and neuronal viability (NeuN). Cerebral glucose metabolism was evaluated using [1] [8]F-FDG positron emission tomography (PET). ACEA administration reduced tau phosphorylation, glial activation, IL-1β expression, and oxidative stress, while increasing IL-10 levels, neuronal preservation, and cerebral glucose metabolism. AM251 treatment aggravated tau pathology, neuroinflammation, oxidative imbalance, and cognitive impairment. Double immunofluorescence demonstrated CB1 receptor colocalization with both Iba-1-positive microglia and GFAP-positive astrocytes, with CB1 predominantly localized to microglia, suggesting a microglia-dependent mechanism underlying CB1-mediated neuroprotection. Aβ levels were not affected by either treatment. Chronic CB1 receptor activation attenuates tau-associated pathology and metabolic dysfunction in 3xTg-AD mice, indicating the therapeutic relevance of CB1 signaling modulation in neurodegenerative disorders.
Additional Links: PMID-41314098
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41314098,
year = {2025},
author = {Ye, M and Kim, JS and Shim, I},
title = {CB1 receptor activation and inhibition differentially modulate cognitive deficits and neuropathology in 3xTg-AD mice.},
journal = {Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie},
volume = {193},
number = {},
pages = {118818},
doi = {10.1016/j.biopha.2025.118818},
pmid = {41314098},
issn = {1950-6007},
abstract = {Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) deposition, tau hyperphosphorylation, neuroinflammation, oxidative stress, and progressive neuronal loss. The endocannabinoid system regulates synaptic function, inflammation, and redox homeostasis through cannabinoid receptor type 1 (CB1). This study aimed to determine whether pharmacological activation or inhibition of the CB1 receptor differentially modulates Alzheimer's disease-related pathology. 3xTg-AD mice received weekly intraperitoneal injections of the CB1 agonist ACEA (1 mg/kg) or the inverse agonist AM251 (1 mg/kg) from 6 to 12 months of age. Cognitive function was assessed using the Morris Water Maze (MWM) and Y-maze, while hippocampal tissues were analyzed for Aβ, p-Tau, glial markers (GFAP, Iba-1), cytokines (IL-1β, IL-10), oxidative stress markers (SOD, GSH, MDA), and neuronal viability (NeuN). Cerebral glucose metabolism was evaluated using [1] [8]F-FDG positron emission tomography (PET). ACEA administration reduced tau phosphorylation, glial activation, IL-1β expression, and oxidative stress, while increasing IL-10 levels, neuronal preservation, and cerebral glucose metabolism. AM251 treatment aggravated tau pathology, neuroinflammation, oxidative imbalance, and cognitive impairment. Double immunofluorescence demonstrated CB1 receptor colocalization with both Iba-1-positive microglia and GFAP-positive astrocytes, with CB1 predominantly localized to microglia, suggesting a microglia-dependent mechanism underlying CB1-mediated neuroprotection. Aβ levels were not affected by either treatment. Chronic CB1 receptor activation attenuates tau-associated pathology and metabolic dysfunction in 3xTg-AD mice, indicating the therapeutic relevance of CB1 signaling modulation in neurodegenerative disorders.},
}
RevDate: 2025-11-28
EEG-based stratification in Alzheimer's disease: Cognitive progression, pathological marker associations, and therapeutic interventions.
Clinical neurophysiology : official journal of the International Federation of Clinical Neurophysiology, 182:2111440 pii:S1388-2457(25)01292-1 [Epub ahead of print].
OBJECTIVE: Clinical cognitive and pathological marker stratification systems have evolved separately, causing mismatches that limit their clinical use. This study retrospectively validated the link between EEG and clinical symptoms, pathological markers, and the therapeutic efficacy of repetitive transcranial magnetic stimulation (rTMS).
METHODS: This multicenter study involved 308 Alzheimer's patients (AD), 176 with Parkinson's (PD), and 181 normal controls. Resting-state EEG were analyzed to identify four oscillation modes. An EEG-based cognitive risk system was created and validated, also evaluating its effect on rTMS therapy effectiveness.
RESULTS: EEG oscillation changes correlated with cognitive decline, revealing distinct brain network disruptions in AD and PD. These oscillation changes were associated with AD biomarkers, particularly tau hyperphosphorylation. Multicenter validation showed an 83% concordance with the Clinical Dementia Rating Scale, and EEG stratification enhanced rTMS therapeutic efficacy.
CONCLUSIONS: This study showed that EEG-based stratification can assess cognitive function, track disease progression, identify key intervention periods, and improve patient selection for better treatment outcomes in clinical settings.
SIGNIFICANCE: This study demonstrates that EEG can connect disease processes to clinical symptoms at a molecular level, offering a unified framework for improved dementia management. This method allows for dynamic monitoring and precise neuromodulation, enhancing personalized care for neurodegenerative diseases.
Additional Links: PMID-41313999
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313999,
year = {2025},
author = {Pei, Z and Zhu, L and Ren, H and Liu, Y and Shi, X and Lin, Y and Wang, J and Li, P and Wang, P and Ji, Y and Zhou, Y and Tang, X and Jiang, X and Tong, X and Guo, Y},
title = {EEG-based stratification in Alzheimer's disease: Cognitive progression, pathological marker associations, and therapeutic interventions.},
journal = {Clinical neurophysiology : official journal of the International Federation of Clinical Neurophysiology},
volume = {182},
number = {},
pages = {2111440},
doi = {10.1016/j.clinph.2025.2111440},
pmid = {41313999},
issn = {1872-8952},
abstract = {OBJECTIVE: Clinical cognitive and pathological marker stratification systems have evolved separately, causing mismatches that limit their clinical use. This study retrospectively validated the link between EEG and clinical symptoms, pathological markers, and the therapeutic efficacy of repetitive transcranial magnetic stimulation (rTMS).
METHODS: This multicenter study involved 308 Alzheimer's patients (AD), 176 with Parkinson's (PD), and 181 normal controls. Resting-state EEG were analyzed to identify four oscillation modes. An EEG-based cognitive risk system was created and validated, also evaluating its effect on rTMS therapy effectiveness.
RESULTS: EEG oscillation changes correlated with cognitive decline, revealing distinct brain network disruptions in AD and PD. These oscillation changes were associated with AD biomarkers, particularly tau hyperphosphorylation. Multicenter validation showed an 83% concordance with the Clinical Dementia Rating Scale, and EEG stratification enhanced rTMS therapeutic efficacy.
CONCLUSIONS: This study showed that EEG-based stratification can assess cognitive function, track disease progression, identify key intervention periods, and improve patient selection for better treatment outcomes in clinical settings.
SIGNIFICANCE: This study demonstrates that EEG can connect disease processes to clinical symptoms at a molecular level, offering a unified framework for improved dementia management. This method allows for dynamic monitoring and precise neuromodulation, enhancing personalized care for neurodegenerative diseases.},
}
RevDate: 2025-11-28
Discovery of novel harmine derivatives as potent, selective, and brain permeable GSK3β inhibitors with effective In vivo anti-AD activity.
European journal of medicinal chemistry, 303:118389 pii:S0223-5234(25)01154-7 [Epub ahead of print].
Alzheimer's disease (AD) is a neurodegenerative disorder with limited therapeutic options. Glycogen synthase kinase 3β (GSK3β), a key enzyme in tau phosphorylation, is a promising therapeutic target for AD. Herein, we employed a structure-based drug design strategy to develop a novel series of harmine derivatives as potent GSK3β inhibitors. Among them, compound 39a bearing an intramolecular hydrogen bond scaffold, showed potent GSK3β inhibition (IC50 = 0.37 nM), meanwhile maintaining remarkable kinase selectivity, including >25000-fold selectivity over dual specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A), a well-known target of β-carboline derivative harmine. It suppressed tau phosphorylation in Tau (P301L) 293T cells (EC50 = 0.06 ± 0.01 μM) and exhibited favorable blood-brain barrier permeability. Notably, 39a significantly attenuated cognitive deficits and tau hyperphosphorylation pathology in OA-induced C57BL/6J mice and 3 × Tg-AD mouse models, without causing spontaneous locomotor defects at therapeutic doses. Collectively, 39a emerges as a promising GSK3β inhibitor for probing GSK3β's role in AD pathogenesis and guiding anti-AD drug discovery.
Additional Links: PMID-41313964
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313964,
year = {2025},
author = {Liu, W and Wu, L and Zeng, X and Chen, H and Ning, X and Sun, X and Xuan, W and Hao, Y and Jia, H and Li, Z and Xu, Z and Li, X and Xu, Z and Liu, W and Zhao, Q and Hao, S},
title = {Discovery of novel harmine derivatives as potent, selective, and brain permeable GSK3β inhibitors with effective In vivo anti-AD activity.},
journal = {European journal of medicinal chemistry},
volume = {303},
number = {},
pages = {118389},
doi = {10.1016/j.ejmech.2025.118389},
pmid = {41313964},
issn = {1768-3254},
abstract = {Alzheimer's disease (AD) is a neurodegenerative disorder with limited therapeutic options. Glycogen synthase kinase 3β (GSK3β), a key enzyme in tau phosphorylation, is a promising therapeutic target for AD. Herein, we employed a structure-based drug design strategy to develop a novel series of harmine derivatives as potent GSK3β inhibitors. Among them, compound 39a bearing an intramolecular hydrogen bond scaffold, showed potent GSK3β inhibition (IC50 = 0.37 nM), meanwhile maintaining remarkable kinase selectivity, including >25000-fold selectivity over dual specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A), a well-known target of β-carboline derivative harmine. It suppressed tau phosphorylation in Tau (P301L) 293T cells (EC50 = 0.06 ± 0.01 μM) and exhibited favorable blood-brain barrier permeability. Notably, 39a significantly attenuated cognitive deficits and tau hyperphosphorylation pathology in OA-induced C57BL/6J mice and 3 × Tg-AD mouse models, without causing spontaneous locomotor defects at therapeutic doses. Collectively, 39a emerges as a promising GSK3β inhibitor for probing GSK3β's role in AD pathogenesis and guiding anti-AD drug discovery.},
}
RevDate: 2025-11-28
Exploring the selective butyrylcholinesterase inhibition potential of phenol carbamates: Experimental and computational study.
European journal of medicinal chemistry, 302(Pt 3):118375 pii:S0223-5234(25)01140-7 [Epub ahead of print].
A series of fourteen novel phenol carbamates was synthesized and evaluated as potential selective butyrylcholinesterase (BChE) inhibitors targeting cholinergic dysfunction in Alzheimer's disease (AD). The compounds were prepared efficiently from resveratrol analogs via a Wittig reaction followed by carbamoylation, and their structures were confirmed by NMR, MS, and HRMS analyses. All derivatives were screened for inhibitory activity against acetylcholinesterase (AChE) and BChE using a modified Ellman method. None of the compounds inhibited AChE, whereas all selectively inhibited BChE, with IC50 values ranging from 0.045 to 6.840 μM. The most potent inhibitor, compound 13, bearing a pyrrolidine moiety, exhibited an IC50 value of 0.045 μM, outperforming the reference drug galantamine by more than two orders of magnitude. Molecular docking and dynamics simulations confirmed strong π-π and alkyl-π interactions between the ligands and the enzyme's active site, accounting for their high affinity and selectivity. In silico ADME(T) analysis predicted excellent intestinal absorption, blood-brain barrier penetration, and low cytotoxicity, while minor genotoxicity alerts were observed for a few derivatives. In vitro cytotoxicity assays in HepG2 cells confirmed the absence of toxicity at concentrations up to 30 μM. These results highlight methoxy-substituted phenol carbamates, particularly compound 13, as promising lead structures for the design of selective BChE inhibitors and potential therapeutic agents for the treatment of AD.
Additional Links: PMID-41313889
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313889,
year = {2025},
author = {Jelčić, A and Talić, S and Odak, I and Mlakić, M and Lasić, Z and Roca, S and Šagud, I and Bruketa, T and Bosnar, M and Barić, D and Škorić, I},
title = {Exploring the selective butyrylcholinesterase inhibition potential of phenol carbamates: Experimental and computational study.},
journal = {European journal of medicinal chemistry},
volume = {302},
number = {Pt 3},
pages = {118375},
doi = {10.1016/j.ejmech.2025.118375},
pmid = {41313889},
issn = {1768-3254},
abstract = {A series of fourteen novel phenol carbamates was synthesized and evaluated as potential selective butyrylcholinesterase (BChE) inhibitors targeting cholinergic dysfunction in Alzheimer's disease (AD). The compounds were prepared efficiently from resveratrol analogs via a Wittig reaction followed by carbamoylation, and their structures were confirmed by NMR, MS, and HRMS analyses. All derivatives were screened for inhibitory activity against acetylcholinesterase (AChE) and BChE using a modified Ellman method. None of the compounds inhibited AChE, whereas all selectively inhibited BChE, with IC50 values ranging from 0.045 to 6.840 μM. The most potent inhibitor, compound 13, bearing a pyrrolidine moiety, exhibited an IC50 value of 0.045 μM, outperforming the reference drug galantamine by more than two orders of magnitude. Molecular docking and dynamics simulations confirmed strong π-π and alkyl-π interactions between the ligands and the enzyme's active site, accounting for their high affinity and selectivity. In silico ADME(T) analysis predicted excellent intestinal absorption, blood-brain barrier penetration, and low cytotoxicity, while minor genotoxicity alerts were observed for a few derivatives. In vitro cytotoxicity assays in HepG2 cells confirmed the absence of toxicity at concentrations up to 30 μM. These results highlight methoxy-substituted phenol carbamates, particularly compound 13, as promising lead structures for the design of selective BChE inhibitors and potential therapeutic agents for the treatment of AD.},
}
RevDate: 2025-11-28
CmpDate: 2025-11-28
Gut microbial-derived indole-3-propionate improves cognitive function in Alzheimer's disease.
Science advances, 11(48):eadw8410.
Intermittent fasting (IF) offers a potential strategy to counteract Alzheimer's disease (AD) progression. In our 16-week study on AD transgenic mice, IF positively affected cognitive function and reduced amyloid-β (Aβ) accumulation, verifying the IF's role in modulating neuroinflammation. Multiomics integration revealed strong links between IF-induced hippocampal gene expression, gut microbiota, and serum metabolites beneficial for cognition. Indole-3-propionic acid (IPA) emerged as a pivotal microbial metabolite. Blocking its neuronal receptor, pregnane X receptor (PXR), abolished IF's effects. Human data paralleled these findings, showing lower IPA levels in patients with mild cognitive impairment and AD than in controls. IPA supplementation and IPA-producing Clostridium sporogenes reproduced IF's cognitive benefits, whereas PXR blockade in neurons or disruption of IPA synthesis abrogated them. IPA crossed the blood-brain barrier, exhibited potent anti-inflammatory activity, and suppressed Aβ accumulation, essential for neuroprotection. These results underscore microbial metabolites regulated by IF, particularly IPA, as therapeutic candidates for AD, highlighting the critical role of the gut-brain axis in neurodegeneration.
Additional Links: PMID-41313780
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313780,
year = {2025},
author = {Li, L and Jia, M and Yang, C and Zhao, Y and Hu, J and Zhao, Y and Hu, X and Ning, F and Ding, C and Li, Q and Gong, J and Jia, X and Xu, K and Wang, Y and Zhou, S and Deng, L and Shi, L and Chen, X and Liu, X and Liu, Z},
title = {Gut microbial-derived indole-3-propionate improves cognitive function in Alzheimer's disease.},
journal = {Science advances},
volume = {11},
number = {48},
pages = {eadw8410},
doi = {10.1126/sciadv.adw8410},
pmid = {41313780},
issn = {2375-2548},
mesh = {*Alzheimer Disease/metabolism/drug therapy ; *Gastrointestinal Microbiome ; Animals ; Humans ; *Indoles/metabolism/pharmacology ; *Cognition/drug effects ; Mice ; Mice, Transgenic ; Male ; Amyloid beta-Peptides/metabolism ; Disease Models, Animal ; Hippocampus/metabolism ; *Propionates/metabolism/pharmacology ; Cognitive Dysfunction ; Female ; },
abstract = {Intermittent fasting (IF) offers a potential strategy to counteract Alzheimer's disease (AD) progression. In our 16-week study on AD transgenic mice, IF positively affected cognitive function and reduced amyloid-β (Aβ) accumulation, verifying the IF's role in modulating neuroinflammation. Multiomics integration revealed strong links between IF-induced hippocampal gene expression, gut microbiota, and serum metabolites beneficial for cognition. Indole-3-propionic acid (IPA) emerged as a pivotal microbial metabolite. Blocking its neuronal receptor, pregnane X receptor (PXR), abolished IF's effects. Human data paralleled these findings, showing lower IPA levels in patients with mild cognitive impairment and AD than in controls. IPA supplementation and IPA-producing Clostridium sporogenes reproduced IF's cognitive benefits, whereas PXR blockade in neurons or disruption of IPA synthesis abrogated them. IPA crossed the blood-brain barrier, exhibited potent anti-inflammatory activity, and suppressed Aβ accumulation, essential for neuroprotection. These results underscore microbial metabolites regulated by IF, particularly IPA, as therapeutic candidates for AD, highlighting the critical role of the gut-brain axis in neurodegeneration.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/metabolism/drug therapy
*Gastrointestinal Microbiome
Animals
Humans
*Indoles/metabolism/pharmacology
*Cognition/drug effects
Mice
Mice, Transgenic
Male
Amyloid beta-Peptides/metabolism
Disease Models, Animal
Hippocampus/metabolism
*Propionates/metabolism/pharmacology
Cognitive Dysfunction
Female
RevDate: 2025-11-28
CmpDate: 2025-11-28
Neurophysiological signatures of default mode network dysfunction and cognitive decline in Alzheimer's disease.
Science advances, 11(48):eadt8991.
Neural hyperexcitability and network dysfunction are neurophysiological hallmarks of Alzheimer's disease (AD) in animal studies, but their presence and clinical relevance in humans remain poorly understood. We introduce a perturbation-based approach combining transcranial magnetic stimulation and electroencephalography (TMS-EEG), alongside resting-state EEG (rsEEG), to investigate neurophysiological basis of default mode network (DMN) dysfunction in early AD. While rsEEG revealed global neural slowing and disrupted synchrony, these measures reflected widespread changes in brain neurophysiology without network-specific insights. In contrast, TMS-EEG identified network-specific local hyperexcitability in the parietal DMN and disrupted connectivity with frontal DMN regions, which uniquely predicted distinct cognitive impairments and mediated the link between structural brain integrity and cognition. Our findings provide critical insights into how network-specific neurophysiological disruptions contribute to AD-related cognitive dysfunction. Perturbation-based assessments hold promise as potential markers of early detection, disease progression, and target engagement for disease-modifying therapies aiming to restore abnormal neurophysiology in AD.
Additional Links: PMID-41313764
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313764,
year = {2025},
author = {Ozdemir, RA and Passera, B and Fried, PJ and Press, D and Shaughnessy, LW and Buss, S and Shafi, MM},
title = {Neurophysiological signatures of default mode network dysfunction and cognitive decline in Alzheimer's disease.},
journal = {Science advances},
volume = {11},
number = {48},
pages = {eadt8991},
doi = {10.1126/sciadv.adt8991},
pmid = {41313764},
issn = {2375-2548},
mesh = {*Alzheimer Disease/physiopathology ; Humans ; *Cognitive Dysfunction/physiopathology ; Electroencephalography ; Male ; Female ; Transcranial Magnetic Stimulation ; Aged ; *Default Mode Network/physiopathology ; Brain/physiopathology ; *Nerve Net/physiopathology ; Magnetic Resonance Imaging ; },
abstract = {Neural hyperexcitability and network dysfunction are neurophysiological hallmarks of Alzheimer's disease (AD) in animal studies, but their presence and clinical relevance in humans remain poorly understood. We introduce a perturbation-based approach combining transcranial magnetic stimulation and electroencephalography (TMS-EEG), alongside resting-state EEG (rsEEG), to investigate neurophysiological basis of default mode network (DMN) dysfunction in early AD. While rsEEG revealed global neural slowing and disrupted synchrony, these measures reflected widespread changes in brain neurophysiology without network-specific insights. In contrast, TMS-EEG identified network-specific local hyperexcitability in the parietal DMN and disrupted connectivity with frontal DMN regions, which uniquely predicted distinct cognitive impairments and mediated the link between structural brain integrity and cognition. Our findings provide critical insights into how network-specific neurophysiological disruptions contribute to AD-related cognitive dysfunction. Perturbation-based assessments hold promise as potential markers of early detection, disease progression, and target engagement for disease-modifying therapies aiming to restore abnormal neurophysiology in AD.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/physiopathology
Humans
*Cognitive Dysfunction/physiopathology
Electroencephalography
Male
Female
Transcranial Magnetic Stimulation
Aged
*Default Mode Network/physiopathology
Brain/physiopathology
*Nerve Net/physiopathology
Magnetic Resonance Imaging
RevDate: 2025-11-28
CmpDate: 2025-11-28
Microglial CLEC7A restrains amyloid beta plaque pathology in a mouse model of Alzheimer's disease.
Alzheimer's & dementia : the journal of the Alzheimer's Association, 21(11):e70943.
INTRODUCTION: CLEC7A is a surface receptor that is highly upregulated on microglia in many Alzheimer's disease (AD) models. Little is known about the role that microglial CLEC7A signaling plays in AD-related pathogenesis.
METHODS: We utilized an inducible, central nervous system (CNS) macrophage-specific knockout of Clec7a to evaluate the role of CLEC7A in the 5xFAD mouse model of AD at 5 months of age. We used immunofluorescence microscopy, single-nuclei RNA sequencing, along with biochemical assays, to evaluate plaque burden, microglial activity, and neuronal health.
RESULTS: CNS macrophage-targeted deletion of CLEC7A in 5xFAD mice led to a twofold increase in plaque burden, exacerbated neuritic dystrophy, and altered the expression of neuronal health genes, but did not appreciably impact microglial activation, plaque engulfment, or disease-associated microglia acquisition.
DISCUSSION: These findings identify protective roles for CLEC7A in AD-related amyloidosis and suggest that CLEC7A-targeting therapeutics may offer promising strategies for treatment of AD.
HIGHLIGHTS: Conditional loss of CLEC7A in central nervous system (CNS) macrophages of 5xFAD mice results in increased amyloid beta deposition. Loss of CLEC7A does not alter the disease-associated microglia transcriptional program or affect the recruitment of microglia to plaque surfaces. Exacerbation of amyloid deposition with loss of CNS-macrophage CLEC7A is associated with worsened neuronal health highlighted by increased neuritic dystrophy.
Additional Links: PMID-41313642
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313642,
year = {2025},
author = {Mangalmurti, A and Zengeler, KE and Hollis, A and Golden, E and Riddlemoser, G and Lukens, JR},
title = {Microglial CLEC7A restrains amyloid beta plaque pathology in a mouse model of Alzheimer's disease.},
journal = {Alzheimer's & dementia : the journal of the Alzheimer's Association},
volume = {21},
number = {11},
pages = {e70943},
doi = {10.1002/alz.70943},
pmid = {41313642},
issn = {1552-5279},
support = {ADSF-21-816651/ALZ/Alzheimer's Association/United States ; /NH/NIH HHS/United States ; R01AG071996/AG/NIA NIH HHS/United States ; R01AG087406/AG/NIA NIH HHS/United States ; RF1AG078684/AG/NIA NIH HHS/United States ; //Cure Alzheimer's Fund/ ; //The Steven A Newman AD Award/ ; //Rick Sharp Foundation/ ; //Harrison Family Foundation/ ; T32GM007267//Medical Scientist Training Program/ ; T32AI007496//Immunology Training Program/ ; T32AI007496//National Institute of Allergy and Infectious Diseases/ ; T32GM156694/GM/NIGMS NIH HHS/United States ; },
mesh = {Animals ; *Alzheimer Disease/pathology/metabolism/genetics ; *Plaque, Amyloid/pathology/metabolism ; Disease Models, Animal ; *Microglia/metabolism/pathology ; *Lectins, C-Type/metabolism/genetics ; Mice ; Mice, Transgenic ; Mice, Knockout ; Macrophages/metabolism ; Amyloid beta-Peptides/metabolism ; Humans ; Brain/pathology/metabolism ; },
abstract = {INTRODUCTION: CLEC7A is a surface receptor that is highly upregulated on microglia in many Alzheimer's disease (AD) models. Little is known about the role that microglial CLEC7A signaling plays in AD-related pathogenesis.
METHODS: We utilized an inducible, central nervous system (CNS) macrophage-specific knockout of Clec7a to evaluate the role of CLEC7A in the 5xFAD mouse model of AD at 5 months of age. We used immunofluorescence microscopy, single-nuclei RNA sequencing, along with biochemical assays, to evaluate plaque burden, microglial activity, and neuronal health.
RESULTS: CNS macrophage-targeted deletion of CLEC7A in 5xFAD mice led to a twofold increase in plaque burden, exacerbated neuritic dystrophy, and altered the expression of neuronal health genes, but did not appreciably impact microglial activation, plaque engulfment, or disease-associated microglia acquisition.
DISCUSSION: These findings identify protective roles for CLEC7A in AD-related amyloidosis and suggest that CLEC7A-targeting therapeutics may offer promising strategies for treatment of AD.
HIGHLIGHTS: Conditional loss of CLEC7A in central nervous system (CNS) macrophages of 5xFAD mice results in increased amyloid beta deposition. Loss of CLEC7A does not alter the disease-associated microglia transcriptional program or affect the recruitment of microglia to plaque surfaces. Exacerbation of amyloid deposition with loss of CNS-macrophage CLEC7A is associated with worsened neuronal health highlighted by increased neuritic dystrophy.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Alzheimer Disease/pathology/metabolism/genetics
*Plaque, Amyloid/pathology/metabolism
Disease Models, Animal
*Microglia/metabolism/pathology
*Lectins, C-Type/metabolism/genetics
Mice
Mice, Transgenic
Mice, Knockout
Macrophages/metabolism
Amyloid beta-Peptides/metabolism
Humans
Brain/pathology/metabolism
RevDate: 2025-11-28
CmpDate: 2025-11-28
Integrating explainable artificial intelligence with multiomics systems biology and electronic health record data mining for personalized drug repurposing in Alzheimer's disease.
Briefings in bioinformatics, 26(6):.
Alzheimer's disease (AD) is characterized by region- and patient-specific molecular heterogeneity, which hinders therapeutic design. In this study, we introduce PRISM-ML (PRecision-medicine using Interpretable Systems and Multiomics with Machine Learning), an open-source integrated analysis pipeline that combines interpretable machine learning with systems biology and electronic health records data mining to elucidate the molecular diversity of AD and predict promising drug repurposing opportunities. First, we integrated and harmonized transcriptomic (bulk RNA-seq) and genomic (genome-wide association study) data from 2105 brain samples, each with matched data from the same individual (1363 AD patients, 742 controls; 9 tissues), sourced from three independent studies. Random forest classifiers with SHapley Additive exPlanations identified patient-specific biomarkers; unsupervised clustering resolved 36 molecularly distinct subtissues (defined as clusters of samples within a brain tissue that share a specific expression pattern); and gene-gene coexpression networks prioritized 262 high-centrality bottleneck genes as putative regulators of dysregulated pathways. Next, knowledge graph-based drug repurposing predicted six Food and Drug Administration (FDA)-approved drugs that simultaneously target multiple bottleneck genes and multiple AD-relevant pathways. Notably, in a large US de-identified insurance-claims database (n = 364 733), exposure to promethazine, one of the candidate drugs, was associated with a 57%-62% lower incidence of AD versus an active antihistamine comparator (adjusted hazard ratio 0.38; inverse-probability weighted 0.43; both P < .001), providing real-world support for its repurposing potential. In summary, PRISM-ML, as an explainable multiomics analysis pipeline, is readily transferable to other complex diseases, advancing precision medicine.
Additional Links: PMID-41313603
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313603,
year = {2025},
author = {Mottaqi, M and Zhang, P and Xie, L},
title = {Integrating explainable artificial intelligence with multiomics systems biology and electronic health record data mining for personalized drug repurposing in Alzheimer's disease.},
journal = {Briefings in bioinformatics},
volume = {26},
number = {6},
pages = {},
doi = {10.1093/bib/bbaf623},
pmid = {41313603},
issn = {1477-4054},
support = {R01GM122845//National Institute of General Medical Sciences of the National Institute of Health/ ; R01AG057555//National Institute on Aging of the National Institute of Health/ ; R21AG083302//National Institute on Aging of the National Institute of Health/ ; 2226183//National Science Foundation/ ; },
mesh = {*Alzheimer Disease/drug therapy/genetics/metabolism ; Humans ; *Drug Repositioning/methods ; *Electronic Health Records ; *Data Mining/methods ; *Precision Medicine ; *Systems Biology/methods ; *Artificial Intelligence ; Gene Regulatory Networks ; Multiomics ; },
abstract = {Alzheimer's disease (AD) is characterized by region- and patient-specific molecular heterogeneity, which hinders therapeutic design. In this study, we introduce PRISM-ML (PRecision-medicine using Interpretable Systems and Multiomics with Machine Learning), an open-source integrated analysis pipeline that combines interpretable machine learning with systems biology and electronic health records data mining to elucidate the molecular diversity of AD and predict promising drug repurposing opportunities. First, we integrated and harmonized transcriptomic (bulk RNA-seq) and genomic (genome-wide association study) data from 2105 brain samples, each with matched data from the same individual (1363 AD patients, 742 controls; 9 tissues), sourced from three independent studies. Random forest classifiers with SHapley Additive exPlanations identified patient-specific biomarkers; unsupervised clustering resolved 36 molecularly distinct subtissues (defined as clusters of samples within a brain tissue that share a specific expression pattern); and gene-gene coexpression networks prioritized 262 high-centrality bottleneck genes as putative regulators of dysregulated pathways. Next, knowledge graph-based drug repurposing predicted six Food and Drug Administration (FDA)-approved drugs that simultaneously target multiple bottleneck genes and multiple AD-relevant pathways. Notably, in a large US de-identified insurance-claims database (n = 364 733), exposure to promethazine, one of the candidate drugs, was associated with a 57%-62% lower incidence of AD versus an active antihistamine comparator (adjusted hazard ratio 0.38; inverse-probability weighted 0.43; both P < .001), providing real-world support for its repurposing potential. In summary, PRISM-ML, as an explainable multiomics analysis pipeline, is readily transferable to other complex diseases, advancing precision medicine.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/drug therapy/genetics/metabolism
Humans
*Drug Repositioning/methods
*Electronic Health Records
*Data Mining/methods
*Precision Medicine
*Systems Biology/methods
*Artificial Intelligence
Gene Regulatory Networks
Multiomics
RevDate: 2025-11-28
CmpDate: 2025-11-28
Potential Mechanisms of Exercise-Mediated Ferroptosis Regulation in Central Nervous System Diseases.
Molecular neurobiology, 63(1):198.
As a common health-promoting intervention, exercise is widely recommended for patients with central nervous system (CNS) disorders such as stroke, Parkinson's disease (PD), and Alzheimer's disease (AD). In current clinical practice, however, exercise intensity is often determined based on therapist experience, with low-to-moderate intensities typically chosen for safety reasons. Thus, clarifying the underlying mechanisms is essential for developing precise and personalized exercise prescriptions in the future. Evidence shows that exercise regulates various "exerkines" (e.g., BDNF, Nrf2, TNFAIP3, and SLC2A1), which promote neural repair and influence iron metabolism. Ferroptosis-an iron-dependent, programmed cell death distinct from apoptosis, necrosis, and autophagy-is closely associated with the progression and prognosis of many diseases, particularly those affecting the CNS. This review synthesizes current understanding of ferroptosis in stroke, PD, and AD, describes how key exercise parameters (intensity, type-aerobic vs. resistance, and duration) influence ferroptosis, and summarizes preclinical and clinical evidence on exercise-induced ferroptosis modulation in CNS disorders. Our aim is to provide a mechanistic basis for optimizing exercise prescriptions to enhance functional recovery in patients with CNS conditions.
Additional Links: PMID-41313529
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313529,
year = {2025},
author = {Li, X and Guo, Z and Li, Q and Li, C and Zhang, X},
title = {Potential Mechanisms of Exercise-Mediated Ferroptosis Regulation in Central Nervous System Diseases.},
journal = {Molecular neurobiology},
volume = {63},
number = {1},
pages = {198},
pmid = {41313529},
issn = {1559-1182},
mesh = {Humans ; *Ferroptosis/physiology ; *Exercise/physiology ; Animals ; *Central Nervous System Diseases/metabolism/pathology/therapy/physiopathology ; },
abstract = {As a common health-promoting intervention, exercise is widely recommended for patients with central nervous system (CNS) disorders such as stroke, Parkinson's disease (PD), and Alzheimer's disease (AD). In current clinical practice, however, exercise intensity is often determined based on therapist experience, with low-to-moderate intensities typically chosen for safety reasons. Thus, clarifying the underlying mechanisms is essential for developing precise and personalized exercise prescriptions in the future. Evidence shows that exercise regulates various "exerkines" (e.g., BDNF, Nrf2, TNFAIP3, and SLC2A1), which promote neural repair and influence iron metabolism. Ferroptosis-an iron-dependent, programmed cell death distinct from apoptosis, necrosis, and autophagy-is closely associated with the progression and prognosis of many diseases, particularly those affecting the CNS. This review synthesizes current understanding of ferroptosis in stroke, PD, and AD, describes how key exercise parameters (intensity, type-aerobic vs. resistance, and duration) influence ferroptosis, and summarizes preclinical and clinical evidence on exercise-induced ferroptosis modulation in CNS disorders. Our aim is to provide a mechanistic basis for optimizing exercise prescriptions to enhance functional recovery in patients with CNS conditions.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Ferroptosis/physiology
*Exercise/physiology
Animals
*Central Nervous System Diseases/metabolism/pathology/therapy/physiopathology
RevDate: 2025-11-28
CmpDate: 2025-11-28
Every-Other-Day Feeding Prevents the Loss of Parvalbumin-Expressing Neurons in the Cerebral Cortex of Female 5xFAD Mice.
Molecular neurobiology, 63(1):195.
The present study demonstrates the effects of an every-other-day (EOD) feeding regimen on parvalbumin (PV)-expressing interneurons in the cortex of 5xFAD mice, a well-established animal model of Alzheimer's disease (AD). Female 5xFAD mice and their non-transgenic littermates were maintained on either an ad libitum (AL) or EOD feeding regimen throughout the presymptomatic phase of the pathology, with comprehensive immunohistochemical and Western blot analyses performed in 6-month-old animals. In AL-fed 5xFAD mice, significant reductions in PV-expressing interneurons were observed in the retrosplenial granular, parietal, and somatosensory cortices compared to non-transgenic controls, supporting their established vulnerability in AD pathology. This neuronal loss was accompanied by a decline in the levels of brain-derived neurotrophic factor (BDNF), a key neurotrophin essential for cell survival and synaptic plasticity. Remarkably, four months of EOD feeding prevented the Aβ-induced loss of PV interneurons and increased the total protein levels of the BDNF receptor TrkB, suggesting enhanced neurotrophic signaling. However, the benefits of EOD feeding were not uniform across all molecular markers, with EOD-fed 5xFAD mice retaining deficits in phosphorylated CaMKII and CREB-binding protein (CBP) and biochemical analysis of plasma indicating metabolic stress-related effects. Together, these results align with the GABAergic hypofunction hypothesis in AD, underscoring the importance of PV interneuron plasticity in neurodegeneration and cognitive decline. They also suggest that dietary strategies like EOD feeding may offer partial neuroprotective effects during early AD progression, however, complex stress-related impacts of EOD in the modulation of PV neuron function remain to be elucidated. Future studies are also warranted to more carefully explore the long-term translational potential of dietary interventions and the interplay between metabolic stress and amyloid pathology, in order to identify novel therapeutic targets across distinct neuronal populations.
Additional Links: PMID-41313510
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313510,
year = {2025},
author = {Ciric, J and Perovic, M and Milovanovic, N and Polaka, S and Jovanovic Macura, I and Nestorovic, N and Tesic, V},
title = {Every-Other-Day Feeding Prevents the Loss of Parvalbumin-Expressing Neurons in the Cerebral Cortex of Female 5xFAD Mice.},
journal = {Molecular neurobiology},
volume = {63},
number = {1},
pages = {195},
pmid = {41313510},
issn = {1559-1182},
support = {451-03-136/2025-03/200007//Ministry of Science, Technological Development and Innovation of the Republic of Serbia/ ; 24IVPHA1288043//American Heart Association/ ; 23CDA1051814//American Heart Association/ ; },
mesh = {Animals ; *Parvalbumins/metabolism ; Female ; Mice, Transgenic ; *Cerebral Cortex/pathology/metabolism ; Brain-Derived Neurotrophic Factor/metabolism ; Alzheimer Disease/pathology/metabolism ; *Neurons/metabolism/pathology ; Interneurons/metabolism/pathology ; Mice ; Receptor, trkB/metabolism ; Amyloid beta-Peptides/metabolism ; *Feeding Behavior ; },
abstract = {The present study demonstrates the effects of an every-other-day (EOD) feeding regimen on parvalbumin (PV)-expressing interneurons in the cortex of 5xFAD mice, a well-established animal model of Alzheimer's disease (AD). Female 5xFAD mice and their non-transgenic littermates were maintained on either an ad libitum (AL) or EOD feeding regimen throughout the presymptomatic phase of the pathology, with comprehensive immunohistochemical and Western blot analyses performed in 6-month-old animals. In AL-fed 5xFAD mice, significant reductions in PV-expressing interneurons were observed in the retrosplenial granular, parietal, and somatosensory cortices compared to non-transgenic controls, supporting their established vulnerability in AD pathology. This neuronal loss was accompanied by a decline in the levels of brain-derived neurotrophic factor (BDNF), a key neurotrophin essential for cell survival and synaptic plasticity. Remarkably, four months of EOD feeding prevented the Aβ-induced loss of PV interneurons and increased the total protein levels of the BDNF receptor TrkB, suggesting enhanced neurotrophic signaling. However, the benefits of EOD feeding were not uniform across all molecular markers, with EOD-fed 5xFAD mice retaining deficits in phosphorylated CaMKII and CREB-binding protein (CBP) and biochemical analysis of plasma indicating metabolic stress-related effects. Together, these results align with the GABAergic hypofunction hypothesis in AD, underscoring the importance of PV interneuron plasticity in neurodegeneration and cognitive decline. They also suggest that dietary strategies like EOD feeding may offer partial neuroprotective effects during early AD progression, however, complex stress-related impacts of EOD in the modulation of PV neuron function remain to be elucidated. Future studies are also warranted to more carefully explore the long-term translational potential of dietary interventions and the interplay between metabolic stress and amyloid pathology, in order to identify novel therapeutic targets across distinct neuronal populations.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Parvalbumins/metabolism
Female
Mice, Transgenic
*Cerebral Cortex/pathology/metabolism
Brain-Derived Neurotrophic Factor/metabolism
Alzheimer Disease/pathology/metabolism
*Neurons/metabolism/pathology
Interneurons/metabolism/pathology
Mice
Receptor, trkB/metabolism
Amyloid beta-Peptides/metabolism
*Feeding Behavior
RevDate: 2025-11-28
Glymphatic system dysfunction mediates amyloid deposition and cognitive impairment in Alzheimer's disease: a PET/MRI multimodality imaging study.
EJNMMI research pii:10.1186/s13550-025-01339-y [Epub ahead of print].
Additional Links: PMID-41313418
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313418,
year = {2025},
author = {Xue, H and Bi, S and Chen, Z and He, Y and Xu, W and Xu, X and Cui, B and Hu, Y and Qi, Z and Yan, S and Lu, J},
title = {Glymphatic system dysfunction mediates amyloid deposition and cognitive impairment in Alzheimer's disease: a PET/MRI multimodality imaging study.},
journal = {EJNMMI research},
volume = {},
number = {},
pages = {},
doi = {10.1186/s13550-025-01339-y},
pmid = {41313418},
issn = {2191-219X},
support = {82394434//National Natural Science Foundation of China/ ; 82102010//National Natural Science Foundation of China/ ; },
}
RevDate: 2025-11-28
CmpDate: 2025-11-28
Nrf2/Keap1 Signaling Axis in the Brain: Master Regulator of Oxidative Stress in Neurodegenerative and Psychiatric Disorders.
Molecular neurobiology, 63(1):197.
Oxidative stress is a crucial factor in the development of CNS disorders, including neurodegenerative and psychiatric conditions. The Nrf2/Keap1 signaling axis plays a central role in defending against oxidative damage by regulating antioxidant and cytoprotective gene expression. Beyond its antioxidant function, Nrf2 influences neurogenesis, synaptic plasticity, mitochondrial bioenergetics, and glial neuronal interactions, all of which are vital for maintaining neural integrity and cognitive performance. Dysregulation of this pathway through altered dimerization, post-translational modifications, or impaired regulation contributes to the pathophysiology of Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, leading to protein aggregation, mitochondrial dysfunction, and neuroinflammation. Emerging evidence also implicates aberrant Nrf2 activity in psychiatric disorders such as depression, schizophrenia, and bipolar disorder, where redox imbalance and neuroimmune activation disrupt neural function. This review summarizes the molecular structure and regulation of the Nrf2/Keap1 pathway, including basal and stress-induced activation, post-translational modifications, and cross-talk with PI3K/Akt, MAPK, and NF-κB signaling. We highlight cell-type-specific roles of Nrf2 in neurons, astrocytes, and microglia, and the gene expression networks that drive CNS antioxidant and detoxification responses. Recent therapeutic strategies include natural and synthetic Nrf2 activators, gene therapy approaches, and nanotechnology-based delivery systems. While the translational potential of Nrf2-targeted interventions is considerable, challenges remain, including risks of overactivation and oncogenicity, lack of reliable biomarkers, and barriers related to blood-brain barrier permeability, dose, timing, and bioavailability. By integrating advances in neuroscience, pharmacology, and molecular medicine, this review emphasizes the promise of Nrf2 as a unifying therapeutic target across diverse CNS pathologies. Future directions include precision modulation through epigenetic regulation and circRNAs, as well as personalized pharmacotherapy. The Nrf2/Keap1 axis represents a multidisciplinary platform for developing multimodal interventions to preserve brain health in neurodegenerative and psychiatric disorders.
Additional Links: PMID-41313410
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313410,
year = {2025},
author = {Goel, F and Singh, P and Rai, SN and Yadav, DK},
title = {Nrf2/Keap1 Signaling Axis in the Brain: Master Regulator of Oxidative Stress in Neurodegenerative and Psychiatric Disorders.},
journal = {Molecular neurobiology},
volume = {63},
number = {1},
pages = {197},
pmid = {41313410},
issn = {1559-1182},
mesh = {Humans ; *Kelch-Like ECH-Associated Protein 1/metabolism ; *NF-E2-Related Factor 2/metabolism ; *Signal Transduction/physiology ; *Oxidative Stress/physiology ; *Mental Disorders/metabolism/pathology ; Animals ; *Neurodegenerative Diseases/metabolism/pathology ; *Brain/metabolism/pathology ; },
abstract = {Oxidative stress is a crucial factor in the development of CNS disorders, including neurodegenerative and psychiatric conditions. The Nrf2/Keap1 signaling axis plays a central role in defending against oxidative damage by regulating antioxidant and cytoprotective gene expression. Beyond its antioxidant function, Nrf2 influences neurogenesis, synaptic plasticity, mitochondrial bioenergetics, and glial neuronal interactions, all of which are vital for maintaining neural integrity and cognitive performance. Dysregulation of this pathway through altered dimerization, post-translational modifications, or impaired regulation contributes to the pathophysiology of Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, leading to protein aggregation, mitochondrial dysfunction, and neuroinflammation. Emerging evidence also implicates aberrant Nrf2 activity in psychiatric disorders such as depression, schizophrenia, and bipolar disorder, where redox imbalance and neuroimmune activation disrupt neural function. This review summarizes the molecular structure and regulation of the Nrf2/Keap1 pathway, including basal and stress-induced activation, post-translational modifications, and cross-talk with PI3K/Akt, MAPK, and NF-κB signaling. We highlight cell-type-specific roles of Nrf2 in neurons, astrocytes, and microglia, and the gene expression networks that drive CNS antioxidant and detoxification responses. Recent therapeutic strategies include natural and synthetic Nrf2 activators, gene therapy approaches, and nanotechnology-based delivery systems. While the translational potential of Nrf2-targeted interventions is considerable, challenges remain, including risks of overactivation and oncogenicity, lack of reliable biomarkers, and barriers related to blood-brain barrier permeability, dose, timing, and bioavailability. By integrating advances in neuroscience, pharmacology, and molecular medicine, this review emphasizes the promise of Nrf2 as a unifying therapeutic target across diverse CNS pathologies. Future directions include precision modulation through epigenetic regulation and circRNAs, as well as personalized pharmacotherapy. The Nrf2/Keap1 axis represents a multidisciplinary platform for developing multimodal interventions to preserve brain health in neurodegenerative and psychiatric disorders.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Kelch-Like ECH-Associated Protein 1/metabolism
*NF-E2-Related Factor 2/metabolism
*Signal Transduction/physiology
*Oxidative Stress/physiology
*Mental Disorders/metabolism/pathology
Animals
*Neurodegenerative Diseases/metabolism/pathology
*Brain/metabolism/pathology
RevDate: 2025-11-28
NAD+ restores proteostasis through splicing-dependent autophagy.
Autophagy [Epub ahead of print].
Autophagy preserves neuronal integrity by clearing damaged proteins and organelles, but its efficiency declines with aging and neurodegeneration. Depletion of the oxidized form of nicotinamide adenine dinucleotide (NAD[+]) is a hallmark of this decline, yet how metabolic restoration enhances autophagic control has remained obscure. Meanwhile, alternative RNA splicing errors accumulate in aging brains, compromising proteostasis. Here, we identify a metabolic - transcriptional mechanism linking NAD[+] metabolism to autophagic proteostasis through the NAD[+] -EVA1C axis. Cross-species analyses in C. elegans, mice, and human samples reveal that NAD[+] supplementation corrects hundreds of age- or Alzheimer-associated splicing errors, notably restoring balanced expression of EVA1C isoforms. Loss of EVA1C impairs the memory and proteostatic benefits of NAD[+], underscoring its essential role in neuronal resilience. Mechanistically, NAD[+] rebalances EVA1C isoforms that interact with chaperones BAG1 and HSPA/HSP70, reinforcing their network to facilitate chaperone-assisted selective autophagy and proteasomal degradation of misfolded proteins such as MAPT/tau. Thus, NAD[+] restoration coordinates RNA splicing fidelity with downstream proteostatic systems, establishing a metabolic - transcriptional checkpoint for neuronal quality control. This finding expands the paradigm of autophagy regulation, positioning metabolic splice-switching as a crucial mechanism to maintain proteostasis and suggesting new strategies to combat aging-related neurodegenerative diseases.
Additional Links: PMID-41313318
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313318,
year = {2025},
author = {Ai, R and Fang, EF},
title = {NAD+ restores proteostasis through splicing-dependent autophagy.},
journal = {Autophagy},
volume = {},
number = {},
pages = {},
doi = {10.1080/15548627.2025.2596679},
pmid = {41313318},
issn = {1554-8635},
abstract = {Autophagy preserves neuronal integrity by clearing damaged proteins and organelles, but its efficiency declines with aging and neurodegeneration. Depletion of the oxidized form of nicotinamide adenine dinucleotide (NAD[+]) is a hallmark of this decline, yet how metabolic restoration enhances autophagic control has remained obscure. Meanwhile, alternative RNA splicing errors accumulate in aging brains, compromising proteostasis. Here, we identify a metabolic - transcriptional mechanism linking NAD[+] metabolism to autophagic proteostasis through the NAD[+] -EVA1C axis. Cross-species analyses in C. elegans, mice, and human samples reveal that NAD[+] supplementation corrects hundreds of age- or Alzheimer-associated splicing errors, notably restoring balanced expression of EVA1C isoforms. Loss of EVA1C impairs the memory and proteostatic benefits of NAD[+], underscoring its essential role in neuronal resilience. Mechanistically, NAD[+] rebalances EVA1C isoforms that interact with chaperones BAG1 and HSPA/HSP70, reinforcing their network to facilitate chaperone-assisted selective autophagy and proteasomal degradation of misfolded proteins such as MAPT/tau. Thus, NAD[+] restoration coordinates RNA splicing fidelity with downstream proteostatic systems, establishing a metabolic - transcriptional checkpoint for neuronal quality control. This finding expands the paradigm of autophagy regulation, positioning metabolic splice-switching as a crucial mechanism to maintain proteostasis and suggesting new strategies to combat aging-related neurodegenerative diseases.},
}
RevDate: 2025-11-28
Cognitive test completion time as an indicator of early brain atrophy: Findings from the Cognitive Assessment for Dementia, iPad version 2 (CADi2) in healthy adults.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
BackgroundAlthough several cognitive screening tools are available, early-stage detection remains challenging because individuals in prodromal or mild cognitive impairment stages often score within normal ranges. Assessment of response time, rather than test score alone, may offer greater sensitivity to subtle decline.ObjectiveTo examine whether completion time on the Cognitive Assessment for Dementia, iPad version 2 (CADi2), is associated with early brain changes suggestive of preclinical dementia.MethodsIn this cross-sectional study of 511 cognitively unimpaired adults (290 men, 221 women; mean age, 61 years) at the Health Science Center, Japan, participants completed the 10-item CADi2 and underwent structural magnetic resonance imaging. Based on the ceiling effects in total scores, we analyzed associations between total test duration and regional brain atrophy.ResultsCADi2 time was significantly associated with hippocampal atrophy, while total scores were not. We applied a deep survival analysis model using Alzheimer's Disease Neuroimaging Initiative data to estimate 5-year dementia risk. Using a threshold of ≥0.25, receiver operating characteristic analysis showed that CADi2 time (area under the curve, 0.632) outperformed CADi2 (0.554) and Mini-Mental State Examination (0.582) scores. Only CADi2 time showed a significant odds ratio [OR] for high dementia risk (OR = 1.94; 95% confidence interval: 1.12-3.40; p = 0.018) after adjusting for covariates.ConclusionsThese findings indicate that completion time from a cognitive test without ceiling effects is more suitable than score-based measures for evaluating the predementia stage and that combining time and score metrics may further improve stratification in cognitively normal individuals.
Additional Links: PMID-41313317
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313317,
year = {2025},
author = {Iwasa, K and Takamura, M and Ishida, M and Kuroda, Y and Abe, S and Nagai, A},
title = {Cognitive test completion time as an indicator of early brain atrophy: Findings from the Cognitive Assessment for Dementia, iPad version 2 (CADi2) in healthy adults.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251400746},
doi = {10.1177/13872877251400746},
pmid = {41313317},
issn = {1875-8908},
abstract = {BackgroundAlthough several cognitive screening tools are available, early-stage detection remains challenging because individuals in prodromal or mild cognitive impairment stages often score within normal ranges. Assessment of response time, rather than test score alone, may offer greater sensitivity to subtle decline.ObjectiveTo examine whether completion time on the Cognitive Assessment for Dementia, iPad version 2 (CADi2), is associated with early brain changes suggestive of preclinical dementia.MethodsIn this cross-sectional study of 511 cognitively unimpaired adults (290 men, 221 women; mean age, 61 years) at the Health Science Center, Japan, participants completed the 10-item CADi2 and underwent structural magnetic resonance imaging. Based on the ceiling effects in total scores, we analyzed associations between total test duration and regional brain atrophy.ResultsCADi2 time was significantly associated with hippocampal atrophy, while total scores were not. We applied a deep survival analysis model using Alzheimer's Disease Neuroimaging Initiative data to estimate 5-year dementia risk. Using a threshold of ≥0.25, receiver operating characteristic analysis showed that CADi2 time (area under the curve, 0.632) outperformed CADi2 (0.554) and Mini-Mental State Examination (0.582) scores. Only CADi2 time showed a significant odds ratio [OR] for high dementia risk (OR = 1.94; 95% confidence interval: 1.12-3.40; p = 0.018) after adjusting for covariates.ConclusionsThese findings indicate that completion time from a cognitive test without ceiling effects is more suitable than score-based measures for evaluating the predementia stage and that combining time and score metrics may further improve stratification in cognitively normal individuals.},
}
RevDate: 2025-11-28
Underexplored moderating effects of sex in subjective cognitive decline: A systematic review and evidence gap.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
BackgroundSubjective cognitive decline (SCD) is considered an early symptom of Alzheimer's disease (AD) and other types of dementia. Establishing differences between males and females in the presentation and risk factors associated with SCD is critical for utilizing subjective cognitive assessments in prognosticating dementia.ObjectiveWe performed a comprehensive review of studies examining the moderating effect of sex on the association between SCD and relevant physical and/or mental health-related outcomes.MethodsThis study was performed following the PRISMA guidelines. We conducted database search in Medline, Cochrane, Web of Science (WOS), and CINAHL. Primary studies including data of the moderating effect of sex on the association between SCD and different outcomes were selected.ResultsA total of 16 studies were included. We found limited evidence for a moderating effect of sex in SCD. Most of the available literature explored sex differences in SCD for risk of dementia, cognitive performance, competing risk of death, AD biomarkers, basal forebrain resting-state functional connectivity, brain volume, among other health outcomes. Among SCD individuals, females showed increased risk of cognitive decline, dementia and other health outcomes, whereas males showed increased risk of death and longer sickness absence compared to controls.ConclusionsOur comprehensive review denotes a lack of studies directly testing the moderating effect of sex in SCD. The available literature points to sex specific associations between SCD and multiple clinical outcomes. However, in line with the current effort of the SCD initiative, further research is necessary within this emerging topic.
Additional Links: PMID-41313305
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313305,
year = {2025},
author = {Álvarez-Pérez, Y and Duarte-Díaz, A and Molina, Y and Figueroa-Lamas, N and Diaz-Galvan, P and García-Cabello, E and Gonzalez-Burgos, L and Yánes-Pérez, R and Rivero-Santana, A and Olofsson, JK and Barroso, J and Ferreira, D and Perestelo-Pérez, L and Cedres, N},
title = {Underexplored moderating effects of sex in subjective cognitive decline: A systematic review and evidence gap.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251397411},
doi = {10.1177/13872877251397411},
pmid = {41313305},
issn = {1875-8908},
abstract = {BackgroundSubjective cognitive decline (SCD) is considered an early symptom of Alzheimer's disease (AD) and other types of dementia. Establishing differences between males and females in the presentation and risk factors associated with SCD is critical for utilizing subjective cognitive assessments in prognosticating dementia.ObjectiveWe performed a comprehensive review of studies examining the moderating effect of sex on the association between SCD and relevant physical and/or mental health-related outcomes.MethodsThis study was performed following the PRISMA guidelines. We conducted database search in Medline, Cochrane, Web of Science (WOS), and CINAHL. Primary studies including data of the moderating effect of sex on the association between SCD and different outcomes were selected.ResultsA total of 16 studies were included. We found limited evidence for a moderating effect of sex in SCD. Most of the available literature explored sex differences in SCD for risk of dementia, cognitive performance, competing risk of death, AD biomarkers, basal forebrain resting-state functional connectivity, brain volume, among other health outcomes. Among SCD individuals, females showed increased risk of cognitive decline, dementia and other health outcomes, whereas males showed increased risk of death and longer sickness absence compared to controls.ConclusionsOur comprehensive review denotes a lack of studies directly testing the moderating effect of sex in SCD. The available literature points to sex specific associations between SCD and multiple clinical outcomes. However, in line with the current effort of the SCD initiative, further research is necessary within this emerging topic.},
}
RevDate: 2025-11-28
Predictors of a Gabapentinoid-Loop-Diuretic Prescribing Cascade in U.S. Nursing Home Residents.
Journal of the American Geriatrics Society [Epub ahead of print].
BACKGROUND: Gabapentinoid-related peripheral edema may prompt loop diuretic prescribing. Nursing home (NH) residents may be especially prone to this prescribing cascade. We estimated the incidence and identified predictors of the gabapentinoid-loop diuretic prescribing cascade in NHs.
METHODS: We conducted a retrospective cohort study using 2016-2022 Medicare claims linked with Minimum Data Set assessments. We identified residents aged ≥ 66 years who initiated gabapentinoids in NHs and who had no evidence of loop diuretic use, heart failure, or renal insufficiency during the prior 6 months. The outcome was loop diuretic initiation within 90 days of gabapentinoid initiation. Using multivariable Poisson regression models, we estimated adjusted risk ratios (aRR) with 95% robust confidence intervals to identify predictors. We used pooled logistic regression models to examine the relationship between time-varying gabapentinoid dose and loop diuretic initiation risk.
RESULTS: Among 23,544 residents, 994 (4.2%) experienced a prescribing cascade at a median of 36 days (IQR 15-61) after gabapentinoid initiation. Risk was higher with age 86-90 years (aRR = 1.60) or ≥ 91 years (aRR = 1.38); a diagnosis of chronic pain or fibromyalgia (aRR = 1.16), or diabetes (aRR = 1.23); and receipt of potassium-sparing diuretics (aRR = 1.53), thiazide diuretics (aRR = 1.27), or 15 or more unique medications (aRR = 1.18). Higher (versus lower) weekly gabapentin dose during follow-up was associated with a 1.45 times higher prescribing cascade risk over 13 weeks. Those with Alzheimer's Disease and Related Dementias (aRR = 0.79), or moderate (aRR = 0.72) to severe cognitive impairment (aRR = 0.59) had a lower risk versus those with intact cognition.
CONCLUSIONS: Approximately 1 in 20 NH residents who initiate gabapentinoids receives a loop diuretic within 3 months. Potentially modifiable predictors included existing polypharmacy and titrating gabapentinoid doses. NH clinicians should monitor for edema soon after gabapentinoid initiation and consider dose reductions or discontinuation before adding a loop diuretic.
Additional Links: PMID-41313166
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41313166,
year = {2025},
author = {Hayes, KN and Belanger, E and Oganisian, A and Joshi, R and Wang, XJ and Grove, LR and Corcoran, KL and Zullo, AR},
title = {Predictors of a Gabapentinoid-Loop-Diuretic Prescribing Cascade in U.S. Nursing Home Residents.},
journal = {Journal of the American Geriatrics Society},
volume = {},
number = {},
pages = {},
doi = {10.1111/jgs.70219},
pmid = {41313166},
issn = {1532-5415},
support = {RF1AG089541/AG/NIA NIH HHS/United States ; },
abstract = {BACKGROUND: Gabapentinoid-related peripheral edema may prompt loop diuretic prescribing. Nursing home (NH) residents may be especially prone to this prescribing cascade. We estimated the incidence and identified predictors of the gabapentinoid-loop diuretic prescribing cascade in NHs.
METHODS: We conducted a retrospective cohort study using 2016-2022 Medicare claims linked with Minimum Data Set assessments. We identified residents aged ≥ 66 years who initiated gabapentinoids in NHs and who had no evidence of loop diuretic use, heart failure, or renal insufficiency during the prior 6 months. The outcome was loop diuretic initiation within 90 days of gabapentinoid initiation. Using multivariable Poisson regression models, we estimated adjusted risk ratios (aRR) with 95% robust confidence intervals to identify predictors. We used pooled logistic regression models to examine the relationship between time-varying gabapentinoid dose and loop diuretic initiation risk.
RESULTS: Among 23,544 residents, 994 (4.2%) experienced a prescribing cascade at a median of 36 days (IQR 15-61) after gabapentinoid initiation. Risk was higher with age 86-90 years (aRR = 1.60) or ≥ 91 years (aRR = 1.38); a diagnosis of chronic pain or fibromyalgia (aRR = 1.16), or diabetes (aRR = 1.23); and receipt of potassium-sparing diuretics (aRR = 1.53), thiazide diuretics (aRR = 1.27), or 15 or more unique medications (aRR = 1.18). Higher (versus lower) weekly gabapentin dose during follow-up was associated with a 1.45 times higher prescribing cascade risk over 13 weeks. Those with Alzheimer's Disease and Related Dementias (aRR = 0.79), or moderate (aRR = 0.72) to severe cognitive impairment (aRR = 0.59) had a lower risk versus those with intact cognition.
CONCLUSIONS: Approximately 1 in 20 NH residents who initiate gabapentinoids receives a loop diuretic within 3 months. Potentially modifiable predictors included existing polypharmacy and titrating gabapentinoid doses. NH clinicians should monitor for edema soon after gabapentinoid initiation and consider dose reductions or discontinuation before adding a loop diuretic.},
}
RevDate: 2025-11-28
Glutathione and TRPM2 Inhibition Reduce Amyloid-Beta and Lipopolysaccharide-Induced Apoptosis, Inflammation, and Oxidative Stress in Microglial Cells.
Cell biology international [Epub ahead of print].
Microglia cells impacted by inflammation and Alzheimer's disease produce toxic reactive oxygen species (ROS), emit signaling molecules, and death as a result of microglia being active due to excessive Ca[2+] entering the cells. The TRPM2 channel plays a crucial role in Ca[2+] permeability, inflammation, ROS, and apoptosis changes in the BV2 microglia cells, while glutathione (GSH) treatment reduces the changes through TRPM2 inhibition. However, the effect of TRPM2 inhibitors and GSH treatment on oxidative stress, inflammation, and apoptotic values in BV2 microglia cells activated with LPS and amyloid-beta (Aβ) has not been investigated yet. The study aimed to assess the effects of TRPM2 inhibition and GSH treatment on the values in BV2 cells activated with LPS and Aβ. BV2 cells were divided into five groups: control (CNT), LPS, Aβ, Aβ + LPS, and Aβ + LPS + GSH. Increased levels of inflammation biomarkers (TNF-α, IL-1β, and IL-6), intracellular Ca[2+] level, cytosolic ROS, mitochondrial membrane dysfunction, cell death, apoptosis, caspases (caspase-3, -8, and -9), and TRPM2 current density were observed in the cells stimulated with LPS and Aβ. These values increased more when LPS and Aβ were incubated together. However, these apoptotic, inflammatory, and oxidant levels decreased in cells treated with GSH and TRPM2 blockers. In conclusion, the involvement of TRPM2 stimulation was demonstrated on Aβ and LPS-induced Ca[2+] entry, oxidative stress, inflammation, and apoptosis parameters in microglia cells. TRPM2 inhibition by GSH treatment seems to be a potential source for the prevention of Aβ and LPS-induced oxidative stress, apoptosis, and inflammation.
Additional Links: PMID-41312814
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41312814,
year = {2025},
author = {Akpınar, O and Nazıroğlu, M},
title = {Glutathione and TRPM2 Inhibition Reduce Amyloid-Beta and Lipopolysaccharide-Induced Apoptosis, Inflammation, and Oxidative Stress in Microglial Cells.},
journal = {Cell biology international},
volume = {},
number = {},
pages = {},
doi = {10.1002/cbin.70109},
pmid = {41312814},
issn = {1095-8355},
support = {//This study was supported by the Scientific Research Projects Coordination Unit (BAP) of Süleyman Demirel University (project number: TDK-2021-8387). Project owner was Mustafa Nazıroğlu./ ; },
abstract = {Microglia cells impacted by inflammation and Alzheimer's disease produce toxic reactive oxygen species (ROS), emit signaling molecules, and death as a result of microglia being active due to excessive Ca[2+] entering the cells. The TRPM2 channel plays a crucial role in Ca[2+] permeability, inflammation, ROS, and apoptosis changes in the BV2 microglia cells, while glutathione (GSH) treatment reduces the changes through TRPM2 inhibition. However, the effect of TRPM2 inhibitors and GSH treatment on oxidative stress, inflammation, and apoptotic values in BV2 microglia cells activated with LPS and amyloid-beta (Aβ) has not been investigated yet. The study aimed to assess the effects of TRPM2 inhibition and GSH treatment on the values in BV2 cells activated with LPS and Aβ. BV2 cells were divided into five groups: control (CNT), LPS, Aβ, Aβ + LPS, and Aβ + LPS + GSH. Increased levels of inflammation biomarkers (TNF-α, IL-1β, and IL-6), intracellular Ca[2+] level, cytosolic ROS, mitochondrial membrane dysfunction, cell death, apoptosis, caspases (caspase-3, -8, and -9), and TRPM2 current density were observed in the cells stimulated with LPS and Aβ. These values increased more when LPS and Aβ were incubated together. However, these apoptotic, inflammatory, and oxidant levels decreased in cells treated with GSH and TRPM2 blockers. In conclusion, the involvement of TRPM2 stimulation was demonstrated on Aβ and LPS-induced Ca[2+] entry, oxidative stress, inflammation, and apoptosis parameters in microglia cells. TRPM2 inhibition by GSH treatment seems to be a potential source for the prevention of Aβ and LPS-induced oxidative stress, apoptosis, and inflammation.},
}
RevDate: 2025-11-28
Magnetic resonance imaging eligibility for anti-amyloid monoclonal antibody treatment for Alzheimer disease: a single-centre retrospective review for service planning.
Additional Links: PMID-41312794
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41312794,
year = {2025},
author = {Healy, M and Thomas, S and Brodtmann, A},
title = {Magnetic resonance imaging eligibility for anti-amyloid monoclonal antibody treatment for Alzheimer disease: a single-centre retrospective review for service planning.},
journal = {Internal medicine journal},
volume = {},
number = {},
pages = {},
doi = {10.1111/imj.70254},
pmid = {41312794},
issn = {1445-5994},
}
RevDate: 2025-11-28
Evaluation of a New Methylimidazole-Containing Thiosemicarbazone as a Cu[+]/Cu[2+]-Targeting Ligand in the Context of Alzheimer's Disease.
Chemistry (Weinheim an der Bergstrasse, Germany) [Epub ahead of print].
The binding of copper ions to amyloid-β (Aβ) peptide leads to reactive oxygen species (ROS) formation and toxic soluble oligomers, contributing to oxidative stress in Alzheimer's disease (AD). Thus, studying compounds with moderate copper affinity is a promising strategy to prevent its interaction with Aβ and reduce toxicity. Here, we evaluated a new tri-coordinating thiosemicarbazone (HXE) with chelating properties to regulate cuprotoxicity in AD. The ligand was nontoxic against HT-22 hippocampal neuronal cells and bound Cu[+] and Cu[2+] at pH 7.4, with affinity constants (log Kcond) of 8.7 and 12.3, respectively, showing high selectivity over Zn[2+] (log Kapp = 5.0). In the presence of Aβ and Cu[2+], HXE formed stable ternary complexes at physiological pH. Ascorbate consumption and coumarin-3-carboxylic acid fluorescence assays showed that the ligand significantly reduces Cu(Aβ16)-mediated ROS production. It also prevented Cu[2+]-induced modulation of Aβ40 self-assembly and restored the typical fibrillar structure of apo-Aβ40 aggregates. Overall, HXE effectively modulates metal-associated Aβ toxicity and emerges as a promising candidate for AD bioinorganic management.
Additional Links: PMID-41312650
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41312650,
year = {2025},
author = {Marinho Barbosa, B and Esmieu, C and Galvácsi, A and Costa, MV and Brison, A and Ladeira, SM and de Oliveira, J and Kállay, C and Hureau, C and Rey, NA},
title = {Evaluation of a New Methylimidazole-Containing Thiosemicarbazone as a Cu[+]/Cu[2+]-Targeting Ligand in the Context of Alzheimer's Disease.},
journal = {Chemistry (Weinheim an der Bergstrasse, Germany)},
volume = {},
number = {},
pages = {e02754},
doi = {10.1002/chem.202502754},
pmid = {41312650},
issn = {1521-3765},
support = {ANR-20-CE07-0009//ANR project Copperation/ ; //Conselho Nacional de Desenvolvimento Científico e Tecnológico, Brazil/ ; //Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro/ ; 24/2551-0001978-2//Fundação de Amparo à Pesquisa do Estado do Rio Grande do Sul/ ; 88881.878977/2023-01//Coordenação de Aperfeiçoamento de Pessoal de Nível Superior/ ; },
abstract = {The binding of copper ions to amyloid-β (Aβ) peptide leads to reactive oxygen species (ROS) formation and toxic soluble oligomers, contributing to oxidative stress in Alzheimer's disease (AD). Thus, studying compounds with moderate copper affinity is a promising strategy to prevent its interaction with Aβ and reduce toxicity. Here, we evaluated a new tri-coordinating thiosemicarbazone (HXE) with chelating properties to regulate cuprotoxicity in AD. The ligand was nontoxic against HT-22 hippocampal neuronal cells and bound Cu[+] and Cu[2+] at pH 7.4, with affinity constants (log Kcond) of 8.7 and 12.3, respectively, showing high selectivity over Zn[2+] (log Kapp = 5.0). In the presence of Aβ and Cu[2+], HXE formed stable ternary complexes at physiological pH. Ascorbate consumption and coumarin-3-carboxylic acid fluorescence assays showed that the ligand significantly reduces Cu(Aβ16)-mediated ROS production. It also prevented Cu[2+]-induced modulation of Aβ40 self-assembly and restored the typical fibrillar structure of apo-Aβ40 aggregates. Overall, HXE effectively modulates metal-associated Aβ toxicity and emerges as a promising candidate for AD bioinorganic management.},
}
RevDate: 2025-11-28
Intranasal thermosensitive gel for brain-targeted delivery of α-Cyperone-loaded solid lipid nanoparticles.
Therapeutic delivery [Epub ahead of print].
AIMS: To develop and evaluate α-Cyperone (α-Cy)-loaded solid lipid nanoparticles (SLNs) incorporated into a thermosensitive in situ gel for enhanced intranasal delivery and improved neuroprotective efficacy.
MATERIALS AND METHODS: α-Cy-SLN was prepared using high-pressure homogenization followed by freeze-drying. The formulation was optimized using a Box-Behnken Design, assessing the effects of lipid-to-surfactant ratio on particle size, polydispersity index (PDI), zeta potential, and entrapment efficiency (EE). The optimized SLNs were characterized for physicochemical properties, in vitro drug release, and brain cell penetration.
RESULTS: The optimized α-Cy-SLNs (Run 5) exhibited a mean particle size of 279.5 ± 0.06 nm, PDI of 0.203 ± 0.24, zeta potential of -23.1 ± 2.38 mV, EE of 70.0 ± 2.21%, and product yield of 72.58 ± 1.24%. In vitro studies demonstrated a sustained release of α-Cy from the SLNs, indicating the formulation's potential for prolonged drug delivery. Incorporation into the thermosensitive in situ gel further supported controlled release and enhanced bioavailability.
CONCLUSIONS: The developed α-Cy-SLN in situ gel formulation offers a promising strategy for intranasal delivery, improving α-Cy bioavailability and therapeutic potential for neuroprotection in Alzheimer's disease.
Additional Links: PMID-41312570
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41312570,
year = {2025},
author = {Sonar, Y and Mahajan, D and Kole, E and Daude, R and Naik, J and Bhairav, B},
title = {Intranasal thermosensitive gel for brain-targeted delivery of α-Cyperone-loaded solid lipid nanoparticles.},
journal = {Therapeutic delivery},
volume = {},
number = {},
pages = {1-14},
doi = {10.1080/20415990.2025.2594295},
pmid = {41312570},
issn = {2041-6008},
abstract = {AIMS: To develop and evaluate α-Cyperone (α-Cy)-loaded solid lipid nanoparticles (SLNs) incorporated into a thermosensitive in situ gel for enhanced intranasal delivery and improved neuroprotective efficacy.
MATERIALS AND METHODS: α-Cy-SLN was prepared using high-pressure homogenization followed by freeze-drying. The formulation was optimized using a Box-Behnken Design, assessing the effects of lipid-to-surfactant ratio on particle size, polydispersity index (PDI), zeta potential, and entrapment efficiency (EE). The optimized SLNs were characterized for physicochemical properties, in vitro drug release, and brain cell penetration.
RESULTS: The optimized α-Cy-SLNs (Run 5) exhibited a mean particle size of 279.5 ± 0.06 nm, PDI of 0.203 ± 0.24, zeta potential of -23.1 ± 2.38 mV, EE of 70.0 ± 2.21%, and product yield of 72.58 ± 1.24%. In vitro studies demonstrated a sustained release of α-Cy from the SLNs, indicating the formulation's potential for prolonged drug delivery. Incorporation into the thermosensitive in situ gel further supported controlled release and enhanced bioavailability.
CONCLUSIONS: The developed α-Cy-SLN in situ gel formulation offers a promising strategy for intranasal delivery, improving α-Cy bioavailability and therapeutic potential for neuroprotection in Alzheimer's disease.},
}
RevDate: 2025-11-28
Aging immunity - the role of T and B cells in neurological disorders among older adults.
Neurologia i neurochirurgia polska pii:VM/OJS/J/106498 [Epub ahead of print].
INTRODUCTION: Immunosenescence is a natural process of immune system aging, which leads to significant changes in the functioning of both innate and adaptive immunity. Alterations in T and B lymphocytes can significantly impact the progression of neurological diseases including multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS).
STATE OF THE ART: Immunosenescence affects T and B cell subsets, reducing their proliferative capacity and altering cytokine profiles. In MS, these changes promote disease progression and diminish responses to immunomodulatory therapies. In AD and PD, dysfunctional T and B cells contribute to sustained neuroinflammation, exacerbating neurodegeneration. ALS is similarly associated with altered adaptive immunity.
CLINICAL IMPLICATIONS: Recognizing how immunosenescent T and B cells contribute to disease in older adults is crucial for refining treatment strategies. These age-related immune changes may explain varied responses to therapies and highlight the need for novel approaches targeting the aged immune system in neurodegenerative diseases.
FUTURE DIRECTIONS: Future research should focus on identifying the mechanisms by which immunosenescent lymphocytes modulate neuroinflammation and neurodegeneration in aging populations. Novel biomarkers and immunomodulatory therapies tailored to older adults could significantly improve outcomes in patients with neurological diseases.
Additional Links: PMID-41312566
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41312566,
year = {2025},
author = {Kwinta, R and Morawiec, N and Bączyk, J and Kubicka-Bączyk, K and Adamczyk-Sowa, M},
title = {Aging immunity - the role of T and B cells in neurological disorders among older adults.},
journal = {Neurologia i neurochirurgia polska},
volume = {},
number = {},
pages = {},
doi = {10.5603/pjnns.106498},
pmid = {41312566},
issn = {0028-3843},
abstract = {INTRODUCTION: Immunosenescence is a natural process of immune system aging, which leads to significant changes in the functioning of both innate and adaptive immunity. Alterations in T and B lymphocytes can significantly impact the progression of neurological diseases including multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS).
STATE OF THE ART: Immunosenescence affects T and B cell subsets, reducing their proliferative capacity and altering cytokine profiles. In MS, these changes promote disease progression and diminish responses to immunomodulatory therapies. In AD and PD, dysfunctional T and B cells contribute to sustained neuroinflammation, exacerbating neurodegeneration. ALS is similarly associated with altered adaptive immunity.
CLINICAL IMPLICATIONS: Recognizing how immunosenescent T and B cells contribute to disease in older adults is crucial for refining treatment strategies. These age-related immune changes may explain varied responses to therapies and highlight the need for novel approaches targeting the aged immune system in neurodegenerative diseases.
FUTURE DIRECTIONS: Future research should focus on identifying the mechanisms by which immunosenescent lymphocytes modulate neuroinflammation and neurodegeneration in aging populations. Novel biomarkers and immunomodulatory therapies tailored to older adults could significantly improve outcomes in patients with neurological diseases.},
}
RevDate: 2025-11-28
Identification of Ageing-related Hub Genes in Humans, Mouse and Rat Based on Bioinformatics Analysis.
Annals of neurosciences [Epub ahead of print].
BACKGROUND: Ageing is a natural process observed in all living organisms. It is a complex biological process involving many genes and pathways. As such, ageing in an organism is associated with an increased likelihood of developing various neurological diseases, for example, Alzheimer's disease (AD) and Parkinson's disease (PD).
PURPOSE: Ageing is associated with many complex processes and functions that are highly interconnected. In this study, we identified pivotal nodes or hubs that significantly contribute to an ageing network, including those highly connected nodes within the network that are particularly important, using available bioinformatics tools in humans and other model organisms. Thus, mutating or altering any of these nodes in a network can result in significant changes in the overall functioning of an organism.
METHODS: For this study, the ageing genes of humans and mice were retrieved from the GenAge database, while the ageing genes of rats were retrieved from the Ageing & Age related Diseases present in Rat Genome Database. STRING (version 11.5), an online tool, was used to create the network. Cytoscape (version 3.10.0), an open-source software with an integrated tool called cytoHubba, was used to identify the hubs from the STRING network in humans, mice and rats. The online tool Enrichr was used to test the functional enrichment of hub genes in humans.
RESULTS: TP53, Trp53 and Actb were identified as important genes in the network, contributing significantly to the process of ageing in humans, mice and rats, respectively, along with others in the network.
CONCLUSION: Identification of these hubs from the network of ageing and ageing-associated genes deserves further investigation to advance existing knowledge and to improve our understanding of ageing in humans and other model organisms.
Additional Links: PMID-41312257
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41312257,
year = {2025},
author = {Chaurasiya, M and Prasad, G},
title = {Identification of Ageing-related Hub Genes in Humans, Mouse and Rat Based on Bioinformatics Analysis.},
journal = {Annals of neurosciences},
volume = {},
number = {},
pages = {09727531251381065},
pmid = {41312257},
issn = {0972-7531},
abstract = {BACKGROUND: Ageing is a natural process observed in all living organisms. It is a complex biological process involving many genes and pathways. As such, ageing in an organism is associated with an increased likelihood of developing various neurological diseases, for example, Alzheimer's disease (AD) and Parkinson's disease (PD).
PURPOSE: Ageing is associated with many complex processes and functions that are highly interconnected. In this study, we identified pivotal nodes or hubs that significantly contribute to an ageing network, including those highly connected nodes within the network that are particularly important, using available bioinformatics tools in humans and other model organisms. Thus, mutating or altering any of these nodes in a network can result in significant changes in the overall functioning of an organism.
METHODS: For this study, the ageing genes of humans and mice were retrieved from the GenAge database, while the ageing genes of rats were retrieved from the Ageing & Age related Diseases present in Rat Genome Database. STRING (version 11.5), an online tool, was used to create the network. Cytoscape (version 3.10.0), an open-source software with an integrated tool called cytoHubba, was used to identify the hubs from the STRING network in humans, mice and rats. The online tool Enrichr was used to test the functional enrichment of hub genes in humans.
RESULTS: TP53, Trp53 and Actb were identified as important genes in the network, contributing significantly to the process of ageing in humans, mice and rats, respectively, along with others in the network.
CONCLUSION: Identification of these hubs from the network of ageing and ageing-associated genes deserves further investigation to advance existing knowledge and to improve our understanding of ageing in humans and other model organisms.},
}
RevDate: 2025-11-28
CmpDate: 2025-11-28
Using AI-Based Gait Analysis to Establish a 5-Meter Walk Time Cutoff for Discriminating Alzheimer's Disease.
Cureus, 17(10):e95491.
Introduction A decline in gait function has been reported to occur early in Alzheimer's disease (AD), a major cause of dementia, suggesting that gait analysis may be a useful tool for dementia screening. However, a simple and practical analysis method or clear cutoff values have yet to be established. This study aimed to analyze gait function using the AI-powered smartphone application "Toruto," identify gait indicators characteristic of elderly patients with AD, and propose effective cutoff values for dementia discrimination. Methods A total of 147 participants were included in the study: 86 healthy elderly individuals and 61 elderly patients with AD (102 female patients and 45 male patients). Exclusion criteria included the use of a cane, the presence of pain during walking, or the need for walking assistance. Gait function at a normal walking speed was analyzed by the AI of "Toruto" to assess speed, rhythm, and left-right asymmetry. An unpaired t-test was used to compare the two groups, and cutoff values for dementia discrimination were calculated using receiver operating characteristic (ROC) curve analysis. The significance level was set at p < 0.05. Results The AD group showed a significant decline in gait speed (12.12 versus 3.98 sec/5 m), rhythm (10.92 versus 6.51), and left-right asymmetry (6.34 versus 2.15) compared with the healthy control group (p < 0.05). ROC curve analysis revealed that using a gait speed cutoff value of 5.85 sec/5 m yielded a sensitivity of 96.7% and a specificity of 96.5%. A rhythm cutoff of 7.06 (sensitivity: 80.3%, specificity: 65.1%) and a left-right asymmetry cutoff of 2.25 (sensitivity: 75.4%, specificity: 67.4%) were also effective discriminative indicators. Conclusion Gait analysis using the AI-powered smartphone application "Toruto" is effective for distinguishing Alzheimer's disease. This study demonstrated that cutoff values, such as 5.85 seconds for a 5-meter walk, serve as practical screening indicators for dementia. As an objective biomarker reflecting cognitive decline, gait function is expected to be useful for the early detection of Alzheimer's disease in both community and clinical settings.
Additional Links: PMID-41311763
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41311763,
year = {2025},
author = {Inoue, T and Sawamura, S and Nagai, T and Kohiyama, K and Takenaka, T and Sera, T and Senba, L},
title = {Using AI-Based Gait Analysis to Establish a 5-Meter Walk Time Cutoff for Discriminating Alzheimer's Disease.},
journal = {Cureus},
volume = {17},
number = {10},
pages = {e95491},
pmid = {41311763},
issn = {2168-8184},
abstract = {Introduction A decline in gait function has been reported to occur early in Alzheimer's disease (AD), a major cause of dementia, suggesting that gait analysis may be a useful tool for dementia screening. However, a simple and practical analysis method or clear cutoff values have yet to be established. This study aimed to analyze gait function using the AI-powered smartphone application "Toruto," identify gait indicators characteristic of elderly patients with AD, and propose effective cutoff values for dementia discrimination. Methods A total of 147 participants were included in the study: 86 healthy elderly individuals and 61 elderly patients with AD (102 female patients and 45 male patients). Exclusion criteria included the use of a cane, the presence of pain during walking, or the need for walking assistance. Gait function at a normal walking speed was analyzed by the AI of "Toruto" to assess speed, rhythm, and left-right asymmetry. An unpaired t-test was used to compare the two groups, and cutoff values for dementia discrimination were calculated using receiver operating characteristic (ROC) curve analysis. The significance level was set at p < 0.05. Results The AD group showed a significant decline in gait speed (12.12 versus 3.98 sec/5 m), rhythm (10.92 versus 6.51), and left-right asymmetry (6.34 versus 2.15) compared with the healthy control group (p < 0.05). ROC curve analysis revealed that using a gait speed cutoff value of 5.85 sec/5 m yielded a sensitivity of 96.7% and a specificity of 96.5%. A rhythm cutoff of 7.06 (sensitivity: 80.3%, specificity: 65.1%) and a left-right asymmetry cutoff of 2.25 (sensitivity: 75.4%, specificity: 67.4%) were also effective discriminative indicators. Conclusion Gait analysis using the AI-powered smartphone application "Toruto" is effective for distinguishing Alzheimer's disease. This study demonstrated that cutoff values, such as 5.85 seconds for a 5-meter walk, serve as practical screening indicators for dementia. As an objective biomarker reflecting cognitive decline, gait function is expected to be useful for the early detection of Alzheimer's disease in both community and clinical settings.},
}
RevDate: 2025-11-28
CmpDate: 2025-11-28
Correction: Five-year dementia prediction and decision support system based on real-world data.
Frontiers in aging neuroscience, 17:1719723.
[This corrects the article DOI: 10.3389/fnagi.2025.1670609.].
Additional Links: PMID-41311655
Full Text:
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41311655,
year = {2025},
author = {Exarchos, TP and Dimakopoulos, GA and Lazaros, K and Krokidis, M and Vrahatis, A and Grammenos, G and Avramouli, A and Skolariki, K and Adams, R and Mahairaki, V and Oh, ES and Leoutsakos, J and Rosenberg, PB and Lyketsos, CG and Vlamos, P},
title = {Correction: Five-year dementia prediction and decision support system based on real-world data.},
journal = {Frontiers in aging neuroscience},
volume = {17},
number = {},
pages = {1719723},
doi = {10.3389/fnagi.2025.1719723},
pmid = {41311655},
issn = {1663-4365},
abstract = {[This corrects the article DOI: 10.3389/fnagi.2025.1670609.].},
}
RevDate: 2025-11-28
CmpDate: 2025-11-28
Diagnostic utility and psychometric properties of the Technology Activities of Daily Living Questionnaire (T-ADLQ) in people with non-formally educated with Alzheimer's disease in Lima, Peru.
Frontiers in aging neuroscience, 17:1660345.
OBJECTIVE: To evaluate the discriminative capacity and psychometric properties of the Technology-Activities of Daily Living Questionnaire (T-ADLQ) in distinguishing cognitively unimpaired individuals from those with amnestic mild cognitive impairment (aMCI) and Alzheimer's disease dementia (ADD) in a population with no formal education.
METHODS: This cross-sectional study included individuals with no formal education over 50 years of age residing in Callao, Peru. Participants were classified into three cognitive groups: cognitively unimpaired (CU; n = 64), aMCI (n = 60), and early ADD (n = 63). Functional decline was assessed with the T-ADLQ. Group comparisons were conducted using the chi-square or ANOVA tests, as appropriate. Pearson correlations were used to assess concurrent validity. The reliability of the T-ADLQ was assessed using Cronbach's alpha. Receiver operating characteristic (ROC) curve analyses and area under the curve (AUC) metrics were used to assess the discriminative validity of the measures across the three cognitive groups.
RESULTS: In a sample of 187 illiterate older adults, the T-ADLQ demonstrated excellent internal consistency (Cronbach's α = 0.966) and strong inverse correlations with global and executive cognitive measures (MMSE, RUDAS, IFS). It also showed a moderate positive correlation with PFAQ. ROC analyses revealed excellent discriminative performance of the T-ADLQ. The total score and the instrumental (IADL) and advanced (AADL) domains achieved perfect accuracy in differentiating cognitively unimpaired individuals from those with aMCI or ADD. The basic activities of daily living (BADL) domain also showed high accuracy, particularly in distinguishing aMCI from ADD.
CONCLUSION: The T-ADLQ and its subdomains exhibit strong psychometric properties and high discriminative capacity in detecting functional decline in individuals with aMCI and ADD. These findings support the T-ADLQ as a valid and reliable tool for assessing functional impairment in populations with no formal education.
Additional Links: PMID-41311654
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41311654,
year = {2025},
author = {Montesinos, R and Olavarria, L and Henriquez, F and Chambergo-Michilot, D and Custodio, B and Delgado, C and Malaga, M and Custodio, N and Slachevsky, A},
title = {Diagnostic utility and psychometric properties of the Technology Activities of Daily Living Questionnaire (T-ADLQ) in people with non-formally educated with Alzheimer's disease in Lima, Peru.},
journal = {Frontiers in aging neuroscience},
volume = {17},
number = {},
pages = {1660345},
pmid = {41311654},
issn = {1663-4365},
abstract = {OBJECTIVE: To evaluate the discriminative capacity and psychometric properties of the Technology-Activities of Daily Living Questionnaire (T-ADLQ) in distinguishing cognitively unimpaired individuals from those with amnestic mild cognitive impairment (aMCI) and Alzheimer's disease dementia (ADD) in a population with no formal education.
METHODS: This cross-sectional study included individuals with no formal education over 50 years of age residing in Callao, Peru. Participants were classified into three cognitive groups: cognitively unimpaired (CU; n = 64), aMCI (n = 60), and early ADD (n = 63). Functional decline was assessed with the T-ADLQ. Group comparisons were conducted using the chi-square or ANOVA tests, as appropriate. Pearson correlations were used to assess concurrent validity. The reliability of the T-ADLQ was assessed using Cronbach's alpha. Receiver operating characteristic (ROC) curve analyses and area under the curve (AUC) metrics were used to assess the discriminative validity of the measures across the three cognitive groups.
RESULTS: In a sample of 187 illiterate older adults, the T-ADLQ demonstrated excellent internal consistency (Cronbach's α = 0.966) and strong inverse correlations with global and executive cognitive measures (MMSE, RUDAS, IFS). It also showed a moderate positive correlation with PFAQ. ROC analyses revealed excellent discriminative performance of the T-ADLQ. The total score and the instrumental (IADL) and advanced (AADL) domains achieved perfect accuracy in differentiating cognitively unimpaired individuals from those with aMCI or ADD. The basic activities of daily living (BADL) domain also showed high accuracy, particularly in distinguishing aMCI from ADD.
CONCLUSION: The T-ADLQ and its subdomains exhibit strong psychometric properties and high discriminative capacity in detecting functional decline in individuals with aMCI and ADD. These findings support the T-ADLQ as a valid and reliable tool for assessing functional impairment in populations with no formal education.},
}
RevDate: 2025-11-28
Advancing Alzheimer's research by improving disease modeling of secondary tauopathy.
NPJ dementia, 1(1):38.
Despite decades of mechanistic investigation of Alzheimer's disease (AD), wide gaps exist in disease modeling, particularly the pathobiological arm of tau pathology. Relying on transgenic models expressing mutated forms of tau has contributed much knowledge about primary tauopathy, yet with limited relevance to human AD. To eliminate blind spots for basic and translational research, we review recent developments in this area and discuss key refinements toward next-generation AD-relevant tauopathy modeling.
Additional Links: PMID-41311528
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41311528,
year = {2025},
author = {Roy, ER and Cao, W},
title = {Advancing Alzheimer's research by improving disease modeling of secondary tauopathy.},
journal = {NPJ dementia},
volume = {1},
number = {1},
pages = {38},
pmid = {41311528},
issn = {3005-1940},
abstract = {Despite decades of mechanistic investigation of Alzheimer's disease (AD), wide gaps exist in disease modeling, particularly the pathobiological arm of tau pathology. Relying on transgenic models expressing mutated forms of tau has contributed much knowledge about primary tauopathy, yet with limited relevance to human AD. To eliminate blind spots for basic and translational research, we review recent developments in this area and discuss key refinements toward next-generation AD-relevant tauopathy modeling.},
}
RevDate: 2025-11-28
CmpDate: 2025-11-28
Sorafenib promotes the E3 ubiquitin ligase FBXW7 to increase tau degradation and ameliorate tauopathies.
Acta pharmaceutica Sinica. B, 15(11):5817-5831.
Tauopathies, including Alzheimer's disease (AD), are a series of neurodegenerative diseases characterized by pathological accumulation of the microtubule-associated protein tau. Since the abnormal modification and deposition of tau in nerve cells are crucial for tauopathy etiology, methods for reducing tau levels, such as promoting tau degradation, may become effective strategies for disease treatment. Herein, we identified that sorafenib significantly reduced total tau and phosphorylated tau levels through screening FDA-approved drugs. We showed that sorafenib treatment attenuated cognitive deficits and tau pathologies in PS19 tauopathy model mice. Mechanistically, we found that sorafenib inhibited multiple kinases involved in tau phosphorylation and promoted autophagy. Importantly, we further demonstrated that sorafenib also promoted the expression of the E3 ubiquitin ligase FBXW7, which could bind tau and mediate tau degradation through the ubiquitin-proteasome pathway. Finally, we showed that FBXW7 expression decreased in the brains of AD patients and tauopathy model mice, and that overexpression of FBXW7 in the hippocampus attenuated cognitive deficits and tau pathologies in PS19 mice. These results suggest that sorafenib may be a promising treatment option for tauopathies by promoting tau degradation and reducing tau phosphorylation, and that targeting FBXW7 could also serve as an alternative therapeutic strategy for tauopathies.
Additional Links: PMID-41311387
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41311387,
year = {2025},
author = {Zhou, Y and Wang, Y and Yang, H and Zhang, C and Meng, J and Zhang, L and Li, K and Huang, LL and Zhang, X and Luo, H and Zhang, Y},
title = {Sorafenib promotes the E3 ubiquitin ligase FBXW7 to increase tau degradation and ameliorate tauopathies.},
journal = {Acta pharmaceutica Sinica. B},
volume = {15},
number = {11},
pages = {5817-5831},
pmid = {41311387},
issn = {2211-3835},
abstract = {Tauopathies, including Alzheimer's disease (AD), are a series of neurodegenerative diseases characterized by pathological accumulation of the microtubule-associated protein tau. Since the abnormal modification and deposition of tau in nerve cells are crucial for tauopathy etiology, methods for reducing tau levels, such as promoting tau degradation, may become effective strategies for disease treatment. Herein, we identified that sorafenib significantly reduced total tau and phosphorylated tau levels through screening FDA-approved drugs. We showed that sorafenib treatment attenuated cognitive deficits and tau pathologies in PS19 tauopathy model mice. Mechanistically, we found that sorafenib inhibited multiple kinases involved in tau phosphorylation and promoted autophagy. Importantly, we further demonstrated that sorafenib also promoted the expression of the E3 ubiquitin ligase FBXW7, which could bind tau and mediate tau degradation through the ubiquitin-proteasome pathway. Finally, we showed that FBXW7 expression decreased in the brains of AD patients and tauopathy model mice, and that overexpression of FBXW7 in the hippocampus attenuated cognitive deficits and tau pathologies in PS19 mice. These results suggest that sorafenib may be a promising treatment option for tauopathies by promoting tau degradation and reducing tau phosphorylation, and that targeting FBXW7 could also serve as an alternative therapeutic strategy for tauopathies.},
}
RevDate: 2025-11-28
CmpDate: 2025-11-28
Electrochemical biosensors with right-side-out-oriented cell membrane coating for the evaluation of AChE inhibitors as potential anti-Alzheimer's disease agents.
Acta pharmaceutica Sinica. B, 15(11):5988-6000.
Biosensors based on acetylcholinesterase (AChE) are crucial for early diagnosis, less invasive treatment, and drug evaluation of Alzheimer's disease (AD). However, existing technologies often suffer from enzyme conformational changes, leading to altered activity and loss and reduced sensor efficacy. To address this challenge, we developed a novel right-side-out-oriented red blood cell membrane-coated electrochemical biosensors (ROCMCBs) to evaluate AChE inhibitors from traditional Chinese medicines (TCMs) as potential anti-AD agents. The developed right-side-out-oriented coating based on immunoaffinity not only fully exposed the binding sites of AChE on the cell membrane but also ensured its conformation and stability as a peripheral membrane-anchoring protein, which was conducive to maintaining its biological activity and producing optimal interaction with drugs. At the same time, the biosensors exhibited a satisfactory sensitivity (limit of detection = 0.41 pmol/L). Ultimately, six potentially active compounds against AD (baicalin, geniposide, gastrodin, berberine, rhynchophylline, and senkyunolide A) were rapidly identified and evaluated from TCMs. This project provides a promising strategy for developing cell membrane-coated electrochemical biosensors. The application of cell membrane-coated electrochemical biosensors with well-defined cell membrane orientation further expands new perspectives and methods for AChE-targeted anti-AD research.
Additional Links: PMID-41311386
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41311386,
year = {2025},
author = {Zhao, Y and Liu, X and Yang, S and Wang, J and Wu, D and Bu, Y and Xie, X},
title = {Electrochemical biosensors with right-side-out-oriented cell membrane coating for the evaluation of AChE inhibitors as potential anti-Alzheimer's disease agents.},
journal = {Acta pharmaceutica Sinica. B},
volume = {15},
number = {11},
pages = {5988-6000},
pmid = {41311386},
issn = {2211-3835},
abstract = {Biosensors based on acetylcholinesterase (AChE) are crucial for early diagnosis, less invasive treatment, and drug evaluation of Alzheimer's disease (AD). However, existing technologies often suffer from enzyme conformational changes, leading to altered activity and loss and reduced sensor efficacy. To address this challenge, we developed a novel right-side-out-oriented red blood cell membrane-coated electrochemical biosensors (ROCMCBs) to evaluate AChE inhibitors from traditional Chinese medicines (TCMs) as potential anti-AD agents. The developed right-side-out-oriented coating based on immunoaffinity not only fully exposed the binding sites of AChE on the cell membrane but also ensured its conformation and stability as a peripheral membrane-anchoring protein, which was conducive to maintaining its biological activity and producing optimal interaction with drugs. At the same time, the biosensors exhibited a satisfactory sensitivity (limit of detection = 0.41 pmol/L). Ultimately, six potentially active compounds against AD (baicalin, geniposide, gastrodin, berberine, rhynchophylline, and senkyunolide A) were rapidly identified and evaluated from TCMs. This project provides a promising strategy for developing cell membrane-coated electrochemical biosensors. The application of cell membrane-coated electrochemical biosensors with well-defined cell membrane orientation further expands new perspectives and methods for AChE-targeted anti-AD research.},
}
RevDate: 2025-11-28
[[11]C](R)-PK11195 Positron Emission Tomography Imaging of Skull Inflammation in Isolated Rapid-Eye-Movement Sleep Behavior Disorder.
Movement disorders : official journal of the Movement Disorder Society [Epub ahead of print].
BACKGROUND: Skull bone marrow is a specialized immune compartment supplying cells to the meninges. Experimental evidence suggests skull marrow activity may contribute to neuroinflammation in early Parkinson's disease, but in vivo assessment has not yet been performed in any α-synucleinopathy.
OBJECTIVE: The objective is to assess skull/meningeal inflammatory activity in patients with isolated rapid-eye-movement sleep behavior disorder (iRBD).
METHODS: 20 iRBD patients recieved longitudinal [[11]C](R)-PK11195 positron emission tomography scans and were compared to 19 healthy controls.
RESULTS: Global skull inflammation in the iRBD patients was 22.87% higher (95% confidence interval: 12.78-32.97) compared to controls, especially in frontal regions, differing from that previously reported in Alzheimer's disease and multiple sclerosis. This inflammation progressed during a 3-year follow-up period but did not correlate with phenoconversion.
CONCLUSION: This is the first in vivo evidence of skull marrow activation in an α-synucleinopathy. Our findings support a role for the skull-meningeal immune axis in iRBD, linking peripheral and central inflammation in prodromal disease. © 2025 International Parkinson and Movement Disorder Society.
Additional Links: PMID-41311354
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41311354,
year = {2025},
author = {Baun, AM and Firbank, M and Hinz, R and Terkelsen, MH and Iranzo, A and Gaig, C and Tolosa, E and Brooks, DJ and Pavese, N and Keogh, M},
title = {[[11]C](R)-PK11195 Positron Emission Tomography Imaging of Skull Inflammation in Isolated Rapid-Eye-Movement Sleep Behavior Disorder.},
journal = {Movement disorders : official journal of the Movement Disorder Society},
volume = {},
number = {},
pages = {},
doi = {10.1002/mds.70133},
pmid = {41311354},
issn = {1531-8257},
support = {CB06/05/0018//Instituto de Salud Carlos III/ ; R4-A104-B60//Danish Parkinson's Association/ ; 4004-00480B//Danmarks Frie Forskningsfond/ ; 8020-00260B//Danmarks Frie Forskningsfond/ ; },
abstract = {BACKGROUND: Skull bone marrow is a specialized immune compartment supplying cells to the meninges. Experimental evidence suggests skull marrow activity may contribute to neuroinflammation in early Parkinson's disease, but in vivo assessment has not yet been performed in any α-synucleinopathy.
OBJECTIVE: The objective is to assess skull/meningeal inflammatory activity in patients with isolated rapid-eye-movement sleep behavior disorder (iRBD).
METHODS: 20 iRBD patients recieved longitudinal [[11]C](R)-PK11195 positron emission tomography scans and were compared to 19 healthy controls.
RESULTS: Global skull inflammation in the iRBD patients was 22.87% higher (95% confidence interval: 12.78-32.97) compared to controls, especially in frontal regions, differing from that previously reported in Alzheimer's disease and multiple sclerosis. This inflammation progressed during a 3-year follow-up period but did not correlate with phenoconversion.
CONCLUSION: This is the first in vivo evidence of skull marrow activation in an α-synucleinopathy. Our findings support a role for the skull-meningeal immune axis in iRBD, linking peripheral and central inflammation in prodromal disease. © 2025 International Parkinson and Movement Disorder Society.},
}
RevDate: 2025-11-28
Brain connectivity as a new target for Alzheimer's disease therapy?.
Brain : a journal of neurology pii:8345027 [Epub ahead of print].
The recent introduction of immunotherapeutic agents targeting amyloid-β (Aβ) has advanced the pharmacological treatment of Alzheimer's disease (AD). Although several anti-Aβ antibodies have dramatically reduced cerebral amyloid plaques, this has not translated into major cognitive or clinical benefits, thus questioning the clinical relevance of these biomarker changes. Indeed, there is an ongoing debate over whether amyloid reduction alone constitutes sufficient evidence of disease modification to justify regulatory approval. Against this backdrop, we propose a third pathway that transcends the binary framework of molecular versus clinical end points by positioning brain connectivity as a system-level intermediate phenotype. This approach is supported by a growing body of evidence. Alterations in brain networks are early, sensitive, and modifiable markers of AD pathology. Connectivity metrics capture the dynamic interplay between genetic and environmental factors, offering a unified model of disease. Advances in precision medicine, such as individualized connectivity 'fingerprints' and the emergence of digital twins, further position brain connectivity as a powerful platform for therapeutic innovation. We argue that adopting brain network analysis as a key outcome measure enables a shift beyond isolated biomarker achievements toward a more integrated, biologically grounded, and clinically meaningful framework for disease modification in AD, bridging the gap between molecular advances and real-world impact.
Additional Links: PMID-41311299
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41311299,
year = {2025},
author = {Pini, L and Allali, G and Imbimbo, BP and Germani, M and Corbetta, M},
title = {Brain connectivity as a new target for Alzheimer's disease therapy?.},
journal = {Brain : a journal of neurology},
volume = {},
number = {},
pages = {},
doi = {10.1093/brain/awaf404},
pmid = {41311299},
issn = {1460-2156},
abstract = {The recent introduction of immunotherapeutic agents targeting amyloid-β (Aβ) has advanced the pharmacological treatment of Alzheimer's disease (AD). Although several anti-Aβ antibodies have dramatically reduced cerebral amyloid plaques, this has not translated into major cognitive or clinical benefits, thus questioning the clinical relevance of these biomarker changes. Indeed, there is an ongoing debate over whether amyloid reduction alone constitutes sufficient evidence of disease modification to justify regulatory approval. Against this backdrop, we propose a third pathway that transcends the binary framework of molecular versus clinical end points by positioning brain connectivity as a system-level intermediate phenotype. This approach is supported by a growing body of evidence. Alterations in brain networks are early, sensitive, and modifiable markers of AD pathology. Connectivity metrics capture the dynamic interplay between genetic and environmental factors, offering a unified model of disease. Advances in precision medicine, such as individualized connectivity 'fingerprints' and the emergence of digital twins, further position brain connectivity as a powerful platform for therapeutic innovation. We argue that adopting brain network analysis as a key outcome measure enables a shift beyond isolated biomarker achievements toward a more integrated, biologically grounded, and clinically meaningful framework for disease modification in AD, bridging the gap between molecular advances and real-world impact.},
}
RevDate: 2025-11-28
CmpDate: 2025-11-28
A scoping review of population-based dementia registries: advancing research, care, and policy.
Alzheimer's & dementia : the journal of the Alzheimer's Association, 21(11):e70938.
Despite their value for public health, research, and care, population-based registries for Alzheimer's disease and related dementias (ADRD) remain limited. We conducted a scoping review of dementia registry studies through December 2023 and identified population-based dementia registries in Organisation for Economic Co-operation and Development countries. We characterized their structure and scope, assessed key themes, and developed recommendations for registry development. We identified 21 population-based dementia registries from a review of 235 publications. These registries help fill gaps in dementia research by providing longitudinal data, improving case identification, and standardizing outcomes for clinical and policy use. However, many registries lack data on healthcare use and caregiving and have limited geographical coverage, thereby reducing their ability to inform research and public health efforts to address dementia burden in an era of rapidly evolving dementia diagnostics and treatments. As dementia cases rise and advancements in prevention, detection, and treatment accelerate, population-based registries are essential for generating real-world evidence to improve dementia care, policy, and outcomes. HIGHLIGHTS: This scoping review identified 21 population-based dementia registries across OECD countries, highlighting the current landscape and structural gaps. Registries provide critical longitudinal data and standardization for research and policy but often lack information on healthcare use, caregiving, and broad geographic representation. With rising dementia rates and evolving treatments, population-based registries are essential for producing real-world evidence to strengthen care, research, and public health planning.
Additional Links: PMID-41311279
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41311279,
year = {2025},
author = {Garland, H and Jacobson, M and Xu, S and Zissimopoulos, J},
title = {A scoping review of population-based dementia registries: advancing research, care, and policy.},
journal = {Alzheimer's & dementia : the journal of the Alzheimer's Association},
volume = {21},
number = {11},
pages = {e70938},
doi = {10.1002/alz.70938},
pmid = {41311279},
issn = {1552-5279},
mesh = {Humans ; *Registries ; *Dementia/therapy/epidemiology ; *Health Policy ; },
abstract = {Despite their value for public health, research, and care, population-based registries for Alzheimer's disease and related dementias (ADRD) remain limited. We conducted a scoping review of dementia registry studies through December 2023 and identified population-based dementia registries in Organisation for Economic Co-operation and Development countries. We characterized their structure and scope, assessed key themes, and developed recommendations for registry development. We identified 21 population-based dementia registries from a review of 235 publications. These registries help fill gaps in dementia research by providing longitudinal data, improving case identification, and standardizing outcomes for clinical and policy use. However, many registries lack data on healthcare use and caregiving and have limited geographical coverage, thereby reducing their ability to inform research and public health efforts to address dementia burden in an era of rapidly evolving dementia diagnostics and treatments. As dementia cases rise and advancements in prevention, detection, and treatment accelerate, population-based registries are essential for generating real-world evidence to improve dementia care, policy, and outcomes. HIGHLIGHTS: This scoping review identified 21 population-based dementia registries across OECD countries, highlighting the current landscape and structural gaps. Registries provide critical longitudinal data and standardization for research and policy but often lack information on healthcare use, caregiving, and broad geographic representation. With rising dementia rates and evolving treatments, population-based registries are essential for producing real-world evidence to strengthen care, research, and public health planning.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Registries
*Dementia/therapy/epidemiology
*Health Policy
RevDate: 2025-11-28
CmpDate: 2025-11-28
[Association between Tau protein deposition and brain metabolites: N-acetylaspartate and creatine as potential biomarkers for advanced Alzheimer's disease].
Nan fang yi ke da xue xue bao = Journal of Southern Medical University, 45(11):2350-2357.
OBJECTIVES: To investigate the associations between Tau protein deposition and brain biochemical metabolites detected by proton magnetic resonance spectroscopy ([1]H-MRS) in patients with advanced Alzheimer's disease (AD).
METHODS: From April, 2022 to December, 2024, 64 Tau-positive AD patients and 29 healthy individuals underwent [18]F-APN-1607 PET/MR and simultaneously acquired multi-voxel [1]H-MRS in the Department of Nuclear Medicine, Nanjing First Hospital. Visual analysis and voxel-based analysis of PET/MR data were performed to investigate the Tau protein deposition patterns in AD patients. Valid voxels within the [1]H-MRS field of view were selected, and their standardized uptake value ratio (SUVr) in PET and metabolite levels of N-acetylaspartate (NAA), choline (Cho), creatine (Cr), NAA/Cr, and Cho/Cr were recorded. The Tau-positive (Tau[+]) voxels and Tau-negative (Tau[-]) voxels of the AD patients were compared for PET and [1]H-MRS parameters, and the correlations between the metabolites and Tau PET SUVr within Tau[+] voxels were analyzed.
RESULTS: Significant Tau protein deposition were observed in the AD patients, involving mainly the bilateral frontal lobes (30.07%), parietal lobes (29.96%), temporal lobes (21.07%), and occipital lobes (15.89%). A total of 1422 valid voxels in AD group (including 994 Tau[+] and 428 Tau[-] voxels) and 814 voxels in the control group were selected. The AD patients showed significantly decreased NAA level and increased SUVr compared with the control group (P<0.05). Subgroup analyses revealed that Tau[+] voxels had higher SUVr and lower Cr and Cho/Cr than Tau[-] voxels (P<0.05). Compared with the control group, Tau[+] voxels exhibited higher SUVr and lower Cr (P<0.05), while Tau[-] voxels showed lower NAA (P=0.004). No significant differences were found in Cho or NAA/Cr among the subgroups (P>0.05). Within Tau[+] voxels, NAA, Cho, and Cr were negatively correlated with SUVr (P<0.001).
CONCLUSIONS: The patients with progressive AD have significant Tau protein deposition in the brain, which is correlated with alterations in metabolite levels. Decreased NAA is more prominent in early or pre-tau deposition stages, while Cr changes is more significant in the regions with Tau protein deposition, suggesting the potential of NAA and Cr as biomarkers for Tau protein deposition in AD for disease monitoring and treatment evaluation.
Additional Links: PMID-41311076
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41311076,
year = {2025},
author = {Li, X and Zhang, Y and Gu, Y and Chen, N and Qian, X and Zhang, P and Hao, J and Wang, F},
title = {[Association between Tau protein deposition and brain metabolites: N-acetylaspartate and creatine as potential biomarkers for advanced Alzheimer's disease].},
journal = {Nan fang yi ke da xue xue bao = Journal of Southern Medical University},
volume = {45},
number = {11},
pages = {2350-2357},
doi = {10.12122/j.issn.1673-4254.2025.11.07},
pmid = {41311076},
issn = {1673-4254},
mesh = {Humans ; *Alzheimer Disease/metabolism/diagnostic imaging ; *Aspartic Acid/analogs & derivatives/metabolism ; *tau Proteins/metabolism ; *Creatine/metabolism ; *Brain/metabolism ; Biomarkers/metabolism ; Positron-Emission Tomography ; Male ; Female ; Proton Magnetic Resonance Spectroscopy ; Choline/metabolism ; Aged ; Middle Aged ; },
abstract = {OBJECTIVES: To investigate the associations between Tau protein deposition and brain biochemical metabolites detected by proton magnetic resonance spectroscopy ([1]H-MRS) in patients with advanced Alzheimer's disease (AD).
METHODS: From April, 2022 to December, 2024, 64 Tau-positive AD patients and 29 healthy individuals underwent [18]F-APN-1607 PET/MR and simultaneously acquired multi-voxel [1]H-MRS in the Department of Nuclear Medicine, Nanjing First Hospital. Visual analysis and voxel-based analysis of PET/MR data were performed to investigate the Tau protein deposition patterns in AD patients. Valid voxels within the [1]H-MRS field of view were selected, and their standardized uptake value ratio (SUVr) in PET and metabolite levels of N-acetylaspartate (NAA), choline (Cho), creatine (Cr), NAA/Cr, and Cho/Cr were recorded. The Tau-positive (Tau[+]) voxels and Tau-negative (Tau[-]) voxels of the AD patients were compared for PET and [1]H-MRS parameters, and the correlations between the metabolites and Tau PET SUVr within Tau[+] voxels were analyzed.
RESULTS: Significant Tau protein deposition were observed in the AD patients, involving mainly the bilateral frontal lobes (30.07%), parietal lobes (29.96%), temporal lobes (21.07%), and occipital lobes (15.89%). A total of 1422 valid voxels in AD group (including 994 Tau[+] and 428 Tau[-] voxels) and 814 voxels in the control group were selected. The AD patients showed significantly decreased NAA level and increased SUVr compared with the control group (P<0.05). Subgroup analyses revealed that Tau[+] voxels had higher SUVr and lower Cr and Cho/Cr than Tau[-] voxels (P<0.05). Compared with the control group, Tau[+] voxels exhibited higher SUVr and lower Cr (P<0.05), while Tau[-] voxels showed lower NAA (P=0.004). No significant differences were found in Cho or NAA/Cr among the subgroups (P>0.05). Within Tau[+] voxels, NAA, Cho, and Cr were negatively correlated with SUVr (P<0.001).
CONCLUSIONS: The patients with progressive AD have significant Tau protein deposition in the brain, which is correlated with alterations in metabolite levels. Decreased NAA is more prominent in early or pre-tau deposition stages, while Cr changes is more significant in the regions with Tau protein deposition, suggesting the potential of NAA and Cr as biomarkers for Tau protein deposition in AD for disease monitoring and treatment evaluation.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alzheimer Disease/metabolism/diagnostic imaging
*Aspartic Acid/analogs & derivatives/metabolism
*tau Proteins/metabolism
*Creatine/metabolism
*Brain/metabolism
Biomarkers/metabolism
Positron-Emission Tomography
Male
Female
Proton Magnetic Resonance Spectroscopy
Choline/metabolism
Aged
Middle Aged
RevDate: 2025-11-28
Elenbecestat and Compound 89 Potently Inhibit BACE1 but Not BACE2 When Subchronically Dosed in Non-Human Primates.
Proteomics [Epub ahead of print].
The β-secretase BACE1 (β-site amyloid precursor (APP) cleaving enzyme 1) is a major drug target for Alzheimer's disease (AD), as it catalyzes the first step in amyloid β (Aβ) generation, but has additional substrates and functions, in particular in the brain. Several advanced clinical trials with BACE1 inhibitors were stopped because of an adverse event, a mild cognitive worsening. The underlying mechanism is not yet known but may result from co-inhibition of the BACE1-homolog BACE2. While a cerebrospinal fluid (CSF) biomarker for measuring BACE2 activity is not yet established, VCAM-1 has been suggested as such a biomarker, but has not yet been tested upon prolonged dosing in vivo. Using CSF pharmacoproteomics and a subchronic dosing paradigm in non-human primates, we demonstrate that compound 89, a BACE inhibitor not yet tested in humans, and the clinically tested drug elenbecestat inhibit BACE1 in vivo, with little or no effect on BACE2, as seen with a reduction of substrates of BACE1, but not of the BACE2 substrate VCAM-1. As a control, verubecestat, which inhibits both BACE2 and BACE1, reduced CSF abundance of BACE1 substrates as well as of VCAM-1. This study demonstrates the suitability of VCAM-1 as a pharmacodynamic biomarker for measuring BACE2 target engagement in CSF.
Additional Links: PMID-41310967
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41310967,
year = {2025},
author = {Tschirner, SK and Schmidt, A and Ito, M and Hyakkoku, K and Yoshimura, A and Müller, SA and Horiguchi, N and Lichtenthaler, SF},
title = {Elenbecestat and Compound 89 Potently Inhibit BACE1 but Not BACE2 When Subchronically Dosed in Non-Human Primates.},
journal = {Proteomics},
volume = {},
number = {},
pages = {e70082},
doi = {10.1002/pmic.70082},
pmid = {41310967},
issn = {1615-9861},
support = {EXC 2145 SyNergy-ID 390857198//Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)/ ; FKZ03LW0246//Federal Ministry of Research, Technology, and Space/ ; SG-23-1029755 BACE/ALZ/Alzheimer's Association/United States ; },
abstract = {The β-secretase BACE1 (β-site amyloid precursor (APP) cleaving enzyme 1) is a major drug target for Alzheimer's disease (AD), as it catalyzes the first step in amyloid β (Aβ) generation, but has additional substrates and functions, in particular in the brain. Several advanced clinical trials with BACE1 inhibitors were stopped because of an adverse event, a mild cognitive worsening. The underlying mechanism is not yet known but may result from co-inhibition of the BACE1-homolog BACE2. While a cerebrospinal fluid (CSF) biomarker for measuring BACE2 activity is not yet established, VCAM-1 has been suggested as such a biomarker, but has not yet been tested upon prolonged dosing in vivo. Using CSF pharmacoproteomics and a subchronic dosing paradigm in non-human primates, we demonstrate that compound 89, a BACE inhibitor not yet tested in humans, and the clinically tested drug elenbecestat inhibit BACE1 in vivo, with little or no effect on BACE2, as seen with a reduction of substrates of BACE1, but not of the BACE2 substrate VCAM-1. As a control, verubecestat, which inhibits both BACE2 and BACE1, reduced CSF abundance of BACE1 substrates as well as of VCAM-1. This study demonstrates the suitability of VCAM-1 as a pharmacodynamic biomarker for measuring BACE2 target engagement in CSF.},
}
RevDate: 2025-11-28
CmpDate: 2025-11-28
Beyond Structure: The Interplay of Bone and Brain During Alzheimer's Disease.
Comprehensive Physiology, 15(6):e70075.
Alzheimer's disease (AD), a leading cause of dementia in the elderly, is traditionally characterized by neurodegeneration driven by amyloid-beta plaques and tau tangles. However, emerging evidence reveals that AD's impact extends beyond the brain, significantly affecting skeletal health. This review integrates clinical and transgenic mouse model data to elucidate the mechanistic interplay between AD pathology and bone metabolism. AD patients exhibit increased risk for hip fractures and low bone mineral density (BMD), independent of cognitive impairment severity. We found altered calcium and alkaline phosphate levels in the blood of patients with mild cognitive impairment and AD, as assessed from the Alzheimer's Disease Neuroimaging Initiative data. Convergent risk factors-age, sex, APOE4 genotype, smoking, and vitamin D deficiency-contribute to both neurodegeneration and bone fragility. Key molecular pathways, such as Wnt/β-catenin signaling and TREM2-mediated osteoclast regulation, underscore shared mechanisms driving disease pathology in both systems. Mouse models of AD consistently demonstrate disrupted bone remodeling, impaired osteoblast function, and heightened osteoclast activity. Therapeutic strategies targeting overlapping pathways, including lithium, anti-FSH antibodies, and NF-κB inhibitors, show promise in mitigating both cognitive decline and bone loss. Collectively, these insights advocate for a more integrated view of AD that includes skeletal comorbidities, potentially guiding the development of dual-purpose interventions.
Additional Links: PMID-41310918
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41310918,
year = {2025},
author = {Pinnamaneni, A and Akkiraju, A and Park, HI and Ch V, RSR and Ayalasomayajula, V and Bandela, M and Kaipa, S and Khosla, S and Zeineh, M and Suryadevara, V and , },
title = {Beyond Structure: The Interplay of Bone and Brain During Alzheimer's Disease.},
journal = {Comprehensive Physiology},
volume = {15},
number = {6},
pages = {e70075},
doi = {10.1002/cph4.70075},
pmid = {41310918},
issn = {2040-4603},
support = {W81XWH-12-2-0012//U.S. Department of Defense/ ; ADNI U01 AG024904/NH/NIH HHS/United States ; },
mesh = {*Alzheimer Disease/metabolism/pathology/physiopathology ; Humans ; Animals ; *Brain/metabolism/pathology ; *Bone and Bones/metabolism/pathology ; Bone Remodeling ; },
abstract = {Alzheimer's disease (AD), a leading cause of dementia in the elderly, is traditionally characterized by neurodegeneration driven by amyloid-beta plaques and tau tangles. However, emerging evidence reveals that AD's impact extends beyond the brain, significantly affecting skeletal health. This review integrates clinical and transgenic mouse model data to elucidate the mechanistic interplay between AD pathology and bone metabolism. AD patients exhibit increased risk for hip fractures and low bone mineral density (BMD), independent of cognitive impairment severity. We found altered calcium and alkaline phosphate levels in the blood of patients with mild cognitive impairment and AD, as assessed from the Alzheimer's Disease Neuroimaging Initiative data. Convergent risk factors-age, sex, APOE4 genotype, smoking, and vitamin D deficiency-contribute to both neurodegeneration and bone fragility. Key molecular pathways, such as Wnt/β-catenin signaling and TREM2-mediated osteoclast regulation, underscore shared mechanisms driving disease pathology in both systems. Mouse models of AD consistently demonstrate disrupted bone remodeling, impaired osteoblast function, and heightened osteoclast activity. Therapeutic strategies targeting overlapping pathways, including lithium, anti-FSH antibodies, and NF-κB inhibitors, show promise in mitigating both cognitive decline and bone loss. Collectively, these insights advocate for a more integrated view of AD that includes skeletal comorbidities, potentially guiding the development of dual-purpose interventions.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/metabolism/pathology/physiopathology
Humans
Animals
*Brain/metabolism/pathology
*Bone and Bones/metabolism/pathology
Bone Remodeling
RevDate: 2025-11-28
CmpDate: 2025-11-28
Paired PET-MRI Deep Learning Model for Translating [[11]C]PiB to [[18]F]Florbetaben Amyloid Images.
Medical physics, 52(12):e70168.
BACKGROUND: Amyloid PET imaging has been extensively employed in the noninvasive assessment of amyloid-beta accumulation in Alzheimer's disease. Various amyloid radiotracers are commonly used in clinical settings; however, the limited interchangeability among these radiotracers hinders the feasibility of long-term clinical trials and multicenter comparisons. The Centiloid method was proposed for standardization, though providing a single score per image; voxel-wise translation remains a formidable task.
PURPOSE: This paper proposes a U-Net model based on a deformable convolution network (DCNv3-based U-Net) for [ 11 C $^{11}\textrm {C}$ ]-Pittsburgh compound B-to-[ 18 F $^{18}\textrm {F}$ ]-florbetaben image translation to augment existing datasets for large-scale model training and provide image information when inconsistencies between visual assessments and the Centiloid scale occur.
METHODS: The DCNv3-based U-Net combined the benefits of deformable convolution that captures long-range dependencies with efficient computation and the encoder-decoder architecture with skip connections for local-global feature learning and image synthesis.
RESULTS: The prediction images presented increased homogeneity to other previous models, closely resembling the texture of [ 18 F $^{18}\textrm {F}$ ]-florbetaben.
CONCLUSIONS: The DCNv3-based U-Net demonstrated high performance in metrics measurement and statistical analyses for the PET image-to-image translation task. This work also justified the importance of MR images in providing structural information.
Additional Links: PMID-41310910
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41310910,
year = {2025},
author = {Tsai, CH and Huang, SY and Lin, YN and Wang, HW and Hsiao, IT and Chen, KT},
title = {Paired PET-MRI Deep Learning Model for Translating [[11]C]PiB to [[18]F]Florbetaben Amyloid Images.},
journal = {Medical physics},
volume = {52},
number = {12},
pages = {e70168},
doi = {10.1002/mp.70168},
pmid = {41310910},
issn = {2473-4209},
support = {NTU-114L7872//Ministry of Education, Taiwan R.O.C./ ; NTU-114L900703//Ministry of Education, Taiwan R.O.C./ ; NTU-113V1015-4//Ministry of Education, Taiwan R.O.C./ ; NTU-114V1015-5//Ministry of Education, Taiwan R.O.C./ ; 114-2628-E-002-019-MY3//National Science and Technology Council/ ; NHRI-EX114- 11205EC//National Health Research Institutes/ ; },
mesh = {*Stilbenes ; *Aniline Compounds ; *Positron-Emission Tomography ; *Magnetic Resonance Imaging ; Humans ; *Deep Learning ; *Image Processing, Computer-Assisted/methods ; *Thiazoles ; *Benzothiazoles ; Carbon Radioisotopes ; *Multimodal Imaging ; *Amyloid/metabolism ; Alzheimer Disease/diagnostic imaging ; *Amyloid beta-Peptides/metabolism ; },
abstract = {BACKGROUND: Amyloid PET imaging has been extensively employed in the noninvasive assessment of amyloid-beta accumulation in Alzheimer's disease. Various amyloid radiotracers are commonly used in clinical settings; however, the limited interchangeability among these radiotracers hinders the feasibility of long-term clinical trials and multicenter comparisons. The Centiloid method was proposed for standardization, though providing a single score per image; voxel-wise translation remains a formidable task.
PURPOSE: This paper proposes a U-Net model based on a deformable convolution network (DCNv3-based U-Net) for [ 11 C $^{11}\
textrm {C}$
]-Pittsburgh compound B-to-[ 18 F $^{18}\
textrm {F}$
]-florbetaben image translation to augment existing datasets for large-scale model training and provide image information when inconsistencies between visual assessments and the Centiloid scale occur.
METHODS: The DCNv3-based U-Net combined the benefits of deformable convolution that captures long-range dependencies with efficient computation and the encoder-decoder architecture with skip connections for local-global feature learning and image synthesis.
RESULTS: The prediction images presented increased homogeneity to other previous models, closely resembling the texture of [ 18 F $^{18}\
textrm {F}$
]-florbetaben.
CONCLUSIONS: The DCNv3-based U-Net demonstrated high performance in metrics measurement and statistical analyses for the PET image-to-image translation task. This work also justified the importance of MR images in providing structural information.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Stilbenes
*Aniline Compounds
*Positron-Emission Tomography
*Magnetic Resonance Imaging
Humans
*Deep Learning
*Image Processing, Computer-Assisted/methods
*Thiazoles
*Benzothiazoles
Carbon Radioisotopes
*Multimodal Imaging
*Amyloid/metabolism
Alzheimer Disease/diagnostic imaging
*Amyloid beta-Peptides/metabolism
RevDate: 2025-11-28
Transferrin receptor-binding blood-brain barrier shuttle enhances brain delivery and plaque-clearing efficacy of a therapeutic anti-Aβ antibody.
Fluids and barriers of the CNS, 22(1):121.
Additional Links: PMID-41310626
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41310626,
year = {2025},
author = {Vega, MR and Hansen, HH and Jensen, CS and Alexiou, E and Madsen, MR and Wichern, F and Kristiansen, MS and Skytte, JL and Salinas, CG and Sotty, F and Jensen, A and Vergo, S and Hecksher-Sørensen, J},
title = {Transferrin receptor-binding blood-brain barrier shuttle enhances brain delivery and plaque-clearing efficacy of a therapeutic anti-Aβ antibody.},
journal = {Fluids and barriers of the CNS},
volume = {22},
number = {1},
pages = {121},
pmid = {41310626},
issn = {2045-8118},
}
RevDate: 2025-11-28
Association between reproductive history, white matter integrity, and cognition in post-menopausal Latina women from the Boston Latino Aging Study.
Journal of clinical and experimental neuropsychology [Epub ahead of print].
BACKGROUND: Women face disproportionately high Alzheimer's disease (AD) rates, with Latina women experiencing particularly elevated cognitive impairment rates. Understanding reproductive factors' impact on brain aging is critical for this underrepresented population. Given sex disparities in AD and reproductive factor influence on brain aging, we examined relations among reproductive history, white matter integrity, and cognitive function in post-menopausal Latina women.
METHODS: Participants were 95 post-menopausal Latina women from the Boston Latino Aging Study, mean age 65.7 (SD = 6.5) years and 11.7 (SD = 4.8) years of education. Reproductive history was obtained via self-report questionnaire. Cognitive assessment included Mini-Mental State Examination, Free and Cued Selective Reminding Test, Category Fluency, Story Memory Delayed Recall, and Symbol Digit Modalities Test administered in Spanish or Portuguese. White matter microstructure was assessed with diffusion weighted imaging using fixel-based methods. Regression models tested associations among reproductive factors, cognitive and imaging outcomes, adjusting for age, education, and cardiovascular risk. Mediation analyses evaluated whether white matter abnormalities explained reproductive-cognitive relations.
RESULTS: Pregnancy history was associated with worse delayed recall, with women having 1-2 (β = -0.79, p = .016) and 3-4 term pregnancies (β = -0.93, p = .005) performing worse than nulliparous women. Overall, hormone replacement therapy use was associated with better delayed recall (β = 0.58, p = .037). White matter analyses revealed trends suggesting pregnancy-related reductions in fiber density and cross-section across multiple tracts, with 3-4 term pregnancies showing most consistent patterns. Hysterectomy showed trends toward higher fiber density in several tracts. Mediation analyses indicated white matter integrity did not account for reproductive-cognitive associations.
CONCLUSIONS: Reproductive history, particularly pregnancy number, is associated with cognitive performance and white matter microstructure in post-menopausal Latina women. These findings underscore the importance of considering reproductive factors in AD risk assessment and highlight the need for longitudinal studies to clarify mechanisms driving these associations.
Additional Links: PMID-41310442
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41310442,
year = {2025},
author = {Giudicessi, A and Koops, E and Vila-Castelar, C and Martinez, L and Ramirez-Perez, N and Silva, G and Medrano, R and Martinez, JE and Solis, I and Alcina, J and Tristão-Pereira, C and Gonzalez Catalan, M and Saldana, DG and Ramírez-Gómez, L and Cronin-Golomb, A and Quiroz, YT},
title = {Association between reproductive history, white matter integrity, and cognition in post-menopausal Latina women from the Boston Latino Aging Study.},
journal = {Journal of clinical and experimental neuropsychology},
volume = {},
number = {},
pages = {1-15},
doi = {10.1080/13803395.2025.2591895},
pmid = {41310442},
issn = {1744-411X},
abstract = {BACKGROUND: Women face disproportionately high Alzheimer's disease (AD) rates, with Latina women experiencing particularly elevated cognitive impairment rates. Understanding reproductive factors' impact on brain aging is critical for this underrepresented population. Given sex disparities in AD and reproductive factor influence on brain aging, we examined relations among reproductive history, white matter integrity, and cognitive function in post-menopausal Latina women.
METHODS: Participants were 95 post-menopausal Latina women from the Boston Latino Aging Study, mean age 65.7 (SD = 6.5) years and 11.7 (SD = 4.8) years of education. Reproductive history was obtained via self-report questionnaire. Cognitive assessment included Mini-Mental State Examination, Free and Cued Selective Reminding Test, Category Fluency, Story Memory Delayed Recall, and Symbol Digit Modalities Test administered in Spanish or Portuguese. White matter microstructure was assessed with diffusion weighted imaging using fixel-based methods. Regression models tested associations among reproductive factors, cognitive and imaging outcomes, adjusting for age, education, and cardiovascular risk. Mediation analyses evaluated whether white matter abnormalities explained reproductive-cognitive relations.
RESULTS: Pregnancy history was associated with worse delayed recall, with women having 1-2 (β = -0.79, p = .016) and 3-4 term pregnancies (β = -0.93, p = .005) performing worse than nulliparous women. Overall, hormone replacement therapy use was associated with better delayed recall (β = 0.58, p = .037). White matter analyses revealed trends suggesting pregnancy-related reductions in fiber density and cross-section across multiple tracts, with 3-4 term pregnancies showing most consistent patterns. Hysterectomy showed trends toward higher fiber density in several tracts. Mediation analyses indicated white matter integrity did not account for reproductive-cognitive associations.
CONCLUSIONS: Reproductive history, particularly pregnancy number, is associated with cognitive performance and white matter microstructure in post-menopausal Latina women. These findings underscore the importance of considering reproductive factors in AD risk assessment and highlight the need for longitudinal studies to clarify mechanisms driving these associations.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-28
Effects of combined group reminiscence and exercise therapy on psychological wellbeing and functional fitness among older adults with dementia.
Scientific reports, 15(1):42449.
Reminiscence therapy and exercise therapy have both proven beneficial for individuals with dementia. However, there is limited information on the effects of combining these two approaches in older adults with dementia. Our study aimed to investigate the impact of combined group reminiscence therapy (GRT) and group exercise therapy (GET) on psychological well-being and functional fitness in this population. A total of 32 older adults with mild to moderate dementia living in care homes were randomly assigned into either intervention or usual care groups. The study was conducted from January to June 2021. Intervention: Participants in intervention group received weekly an hour session of GRT and biweekly 1.25-hour session of GET. Reminiscence therapy was based on Remembering Yesterday and Caring Today module, adapted and modified according to participants' cultural background. GET consisted of stretching, strengthening, aerobic and multicomponent exercises. Outcome measures include the Quality of Life - Alzheimer's Disease (QOL-AD), Addenbrooke's Cognitive Examination-III (ACE-III), Beck Anxiety Inventory (BAI), Satisfaction with Life Scale (SWLS), Geriatric Depression Scale (GDS), and Functional Fitness MOT (FFMOT). Independent sample t-test and Mann-Whitney U test show that the participants from the GRT + GET group reported statistically significant higher quality of life and satisfaction with life, with a medium to large effect size. There are no other statistically significant results found for other psychosocial measures. FFMOT was found to deteriorate in both groups with a lesser amount in the intervention group. This study suggests that combined GRT and GET may induce some psychosocial benefits, in particular quality of life and some positive trend in deceleration of functional fitness deterioration among older adults with mild to moderate dementia. Preserving psychological and physical wellbeing is essential for older adults with dementia to maintain their functional independence for as long as possible.
Additional Links: PMID-41310327
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41310327,
year = {2025},
author = {Subramaniam, P and Singh, DKA and Al-Shahrani, HF and Hammad, MA and Lim, EV and Hoh, XJ and Tay, KW and Alavi, K},
title = {Effects of combined group reminiscence and exercise therapy on psychological wellbeing and functional fitness among older adults with dementia.},
journal = {Scientific reports},
volume = {15},
number = {1},
pages = {42449},
pmid = {41310327},
issn = {2045-2322},
support = {2013-027//Pusat Transformasi Komuniti Universiti , Universiti Putra Malaysia/ ; PNURSP2025R707//Deanship of Scientific Research, Princess Nourah Bint Abdulrahman University/ ; },
mesh = {Humans ; Female ; Male ; Aged ; *Dementia/therapy/psychology ; *Exercise Therapy/methods ; Quality of Life ; Aged, 80 and over ; *Psychotherapy, Group/methods ; Treatment Outcome ; },
abstract = {Reminiscence therapy and exercise therapy have both proven beneficial for individuals with dementia. However, there is limited information on the effects of combining these two approaches in older adults with dementia. Our study aimed to investigate the impact of combined group reminiscence therapy (GRT) and group exercise therapy (GET) on psychological well-being and functional fitness in this population. A total of 32 older adults with mild to moderate dementia living in care homes were randomly assigned into either intervention or usual care groups. The study was conducted from January to June 2021. Intervention: Participants in intervention group received weekly an hour session of GRT and biweekly 1.25-hour session of GET. Reminiscence therapy was based on Remembering Yesterday and Caring Today module, adapted and modified according to participants' cultural background. GET consisted of stretching, strengthening, aerobic and multicomponent exercises. Outcome measures include the Quality of Life - Alzheimer's Disease (QOL-AD), Addenbrooke's Cognitive Examination-III (ACE-III), Beck Anxiety Inventory (BAI), Satisfaction with Life Scale (SWLS), Geriatric Depression Scale (GDS), and Functional Fitness MOT (FFMOT). Independent sample t-test and Mann-Whitney U test show that the participants from the GRT + GET group reported statistically significant higher quality of life and satisfaction with life, with a medium to large effect size. There are no other statistically significant results found for other psychosocial measures. FFMOT was found to deteriorate in both groups with a lesser amount in the intervention group. This study suggests that combined GRT and GET may induce some psychosocial benefits, in particular quality of life and some positive trend in deceleration of functional fitness deterioration among older adults with mild to moderate dementia. Preserving psychological and physical wellbeing is essential for older adults with dementia to maintain their functional independence for as long as possible.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Female
Male
Aged
*Dementia/therapy/psychology
*Exercise Therapy/methods
Quality of Life
Aged, 80 and over
*Psychotherapy, Group/methods
Treatment Outcome
RevDate: 2025-11-27
Plasma biomarkers of alzheimer's disease and related dementias are associated with cognitive change in community-dwelling older individuals in Australia and the US.
GeroScience [Epub ahead of print].
Plasma biomarkers of Alzheimer's disease and related dementias (ADRD) are associated with the risk of dementia. However, the extent to which they could reflect cognitive ageing, and whether this is consistent across factors known to influence biomarkers (e.g. sex and chronic kidney disease) and in different populations, is unknown. Data were from a diverse community-dwelling cohort of older individuals without dementia in Australia (n = 11,930) and the US (n = 1,181). Global cognition, verbal fluency, episodic memory and psychomotor speed were assessed repeatedly over more than a decade. Plasma phosphorylated tau181 (p-tau181), glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL) and amyloid beta (Aβ) 42 and 40 were measured using Simoa technology. Higher levels of p-tau181 (β: -0.001 to -0.212), GFAP (β: -0.022 to -0.300) and NfL (β: -0.022 to -0.219), and lower levels of Aβ42/40 ratio (β: 0.015 to 0.126) were associated with greater cognitive decline over time. Associations were strongest for global cognition, episodic memory, and psychomotor speed, and weaker/non-significant for verbal fluency. All associations were consistent across countries. Furthermore, in stratified analyses, the results did not differ by sex or depending on the presence of chronic kidney disease. We found robust associations between plasma ADRD biomarkers and cognitive change in initially healthy older individuals in both Australia and the US, and across both sexes. Despite chronic kidney disease influencing biomarker levels, associations were consistent among individuals with and without chronic kidney disease. This indicates the potential broad utility of these biomarkers.
Additional Links: PMID-41310263
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41310263,
year = {2025},
author = {Wu, Z and Mielke, MM and Murray, AM and Webb, K and Woods, RL and Owen, A and Orchard, SG and Chong, TT and Sheets, KM and Ryan, J},
title = {Plasma biomarkers of alzheimer's disease and related dementias are associated with cognitive change in community-dwelling older individuals in Australia and the US.},
journal = {GeroScience},
volume = {},
number = {},
pages = {},
pmid = {41310263},
issn = {2509-2723},
support = {U01AG029824/NR/NINR NIH HHS/United States ; U19AG062682/NR/NINR NIH HHS/United States ; 334047//National Health and Medical Research Council/ ; 1127060//National Health and Medical Research Council/ ; 1135727//National Health and Medical Research Council/ ; 1RF1AG079397/AG/NIA NIH HHS/United States ; },
abstract = {Plasma biomarkers of Alzheimer's disease and related dementias (ADRD) are associated with the risk of dementia. However, the extent to which they could reflect cognitive ageing, and whether this is consistent across factors known to influence biomarkers (e.g. sex and chronic kidney disease) and in different populations, is unknown. Data were from a diverse community-dwelling cohort of older individuals without dementia in Australia (n = 11,930) and the US (n = 1,181). Global cognition, verbal fluency, episodic memory and psychomotor speed were assessed repeatedly over more than a decade. Plasma phosphorylated tau181 (p-tau181), glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL) and amyloid beta (Aβ) 42 and 40 were measured using Simoa technology. Higher levels of p-tau181 (β: -0.001 to -0.212), GFAP (β: -0.022 to -0.300) and NfL (β: -0.022 to -0.219), and lower levels of Aβ42/40 ratio (β: 0.015 to 0.126) were associated with greater cognitive decline over time. Associations were strongest for global cognition, episodic memory, and psychomotor speed, and weaker/non-significant for verbal fluency. All associations were consistent across countries. Furthermore, in stratified analyses, the results did not differ by sex or depending on the presence of chronic kidney disease. We found robust associations between plasma ADRD biomarkers and cognitive change in initially healthy older individuals in both Australia and the US, and across both sexes. Despite chronic kidney disease influencing biomarker levels, associations were consistent among individuals with and without chronic kidney disease. This indicates the potential broad utility of these biomarkers.},
}
RevDate: 2025-11-27
Transplantation of GABAergic Interneuron Progenitors Restores Cortical Circuit Function in an Alzheimer's Disease Mouse Model.
Advanced science (Weinheim, Baden-Wurttemberg, Germany) [Epub ahead of print].
In addition to dementia, Alzheimer's patients suffer from sleep impairments and aberrations in sleep-dependent brain rhythms. Deficits in inhibitory GABAergic interneuron function disrupt one of those rhythms, slow oscillation in particular, and actively contribute to Alzheimer's progression. The degree to which transplantation of healthy donor interneuron progenitors restores slow oscillation rhythm in young APP/PS1 mice is tested. Medial ganglionic eminence (MGE) progenitors are harvested from mouse embryos and transplanted them into host APP/PS1 mutant cortices. 3D light-sheet and structured illumination microscopy revealed that transplanted MGE progenitors survived and matured into healthy interneurons. In vivo multiphoton calcium imaging and voltage-sensitive dye imaging showed functional integration and slow oscillation rescue in the absence or presence of optogenetic stimulation. The work provides proof-of-concept evidence that stem cell therapy may serve as a viable strategy to rescue functional impairments in cortical circuits of APP/PS1 mice and potentially those of Alzheimer's patients.
Additional Links: PMID-41310259
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41310259,
year = {2025},
author = {Yokomizo, S and Maci, M and Stafford, AM and Miller, MR and Perle, SJ and Inagaki, S and Takahashi, S and Brown-Harding, H and Liang, L and Lovely, A and Algamal, M and Gillani, RL and Zwang, TJ and Richardson, D and Naegele, JR and Vogt, D and Kastanenka, KV},
title = {Transplantation of GABAergic Interneuron Progenitors Restores Cortical Circuit Function in an Alzheimer's Disease Mouse Model.},
journal = {Advanced science (Weinheim, Baden-Wurttemberg, Germany)},
volume = {},
number = {},
pages = {e11472},
doi = {10.1002/advs.202511472},
pmid = {41310259},
issn = {2198-3844},
support = {R01AG066171/NH/NIH HHS/United States ; R21AG075807/NH/NIH HHS/United States ; //Japan Agency for Medical Research and Development/ ; 202360054//Japan Society for the Promotion of Science/ ; },
abstract = {In addition to dementia, Alzheimer's patients suffer from sleep impairments and aberrations in sleep-dependent brain rhythms. Deficits in inhibitory GABAergic interneuron function disrupt one of those rhythms, slow oscillation in particular, and actively contribute to Alzheimer's progression. The degree to which transplantation of healthy donor interneuron progenitors restores slow oscillation rhythm in young APP/PS1 mice is tested. Medial ganglionic eminence (MGE) progenitors are harvested from mouse embryos and transplanted them into host APP/PS1 mutant cortices. 3D light-sheet and structured illumination microscopy revealed that transplanted MGE progenitors survived and matured into healthy interneurons. In vivo multiphoton calcium imaging and voltage-sensitive dye imaging showed functional integration and slow oscillation rescue in the absence or presence of optogenetic stimulation. The work provides proof-of-concept evidence that stem cell therapy may serve as a viable strategy to rescue functional impairments in cortical circuits of APP/PS1 mice and potentially those of Alzheimer's patients.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-28
Fair positive unlabeled learning for predicting undiagnosed Alzheimer's disease in diverse electronic health records.
NPJ digital medicine, 8(1):730.
Alzheimer's Disease (AD), the most common neurodegenerative disease, is underdiagnosed and more prominent in underrepresented groups. We performed semi-supervised positive unlabeled learning (SSPUL) coupled with racial bias mitigation for equitable prediction of undiagnosed AD from diverse populations at UCLA Health using electronic health records. SSPUL achieved superior sensitivity (0.77-0.81) and area under the precision recall curve (AUCPR) (0.81-0.87) across non-Hispanic white, non-Hispanic African American, Hispanic Latino, and East Asian groups compared to supervised baseline models (sensitivity: 0.39-0.53; AUCPR: 0.3-0.7). SSPUL also exhibited superior fairness as evidenced by the lowest cumulative parity loss. We identified top shared and distinct features among labeled and unlabeled AD patients, including those that are neurological (e.g., memory loss) and non-neurological (e.g., decubitus ulcer). We validated our results using polygenic risk scores, which were higher in labeled and predicted positives than in predicted negatives among non-Hispanic white, Hispanic Latino, and East Asian groups (p < 0.001).
Additional Links: PMID-41310166
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41310166,
year = {2025},
author = {Tran, T and Fu, M and Fung, J and Sankararaman, S and Elashoff, DA and Vossel, K and Chang, TS},
title = {Fair positive unlabeled learning for predicting undiagnosed Alzheimer's disease in diverse electronic health records.},
journal = {NPJ digital medicine},
volume = {8},
number = {1},
pages = {730},
pmid = {41310166},
issn = {2398-6352},
support = {R01AG085518-01A1//National Institute of Aging/ ; R01AG085518-01A1//National Institute of Aging/ ; R01AG085518-01A1//National Institute of Aging/ ; R01AG085518-01A1//National Institute of Aging/ ; R01AG085518-01A1//National Institute of Aging/ ; R01AG085518-01A1//National Institute of Aging/ ; CAREER-1943497//National Science Foundation/ ; R35GM153406/NH/NIH HHS/United States ; R01AG081768A/NH/NIH HHS/United States ; Contract #22-10079//California Department of Public Health, Chronic Disease Control Branch, Alzheimer's Disease Program/ ; Contract #22-10079//California Department of Public Health, Chronic Disease Control Branch, Alzheimer's Disease Program/ ; Contract #22-10079//California Department of Public Health, Chronic Disease Control Branch, Alzheimer's Disease Program/ ; U54NS123746/NS/NINDS NIH HHS/United States ; },
abstract = {Alzheimer's Disease (AD), the most common neurodegenerative disease, is underdiagnosed and more prominent in underrepresented groups. We performed semi-supervised positive unlabeled learning (SSPUL) coupled with racial bias mitigation for equitable prediction of undiagnosed AD from diverse populations at UCLA Health using electronic health records. SSPUL achieved superior sensitivity (0.77-0.81) and area under the precision recall curve (AUCPR) (0.81-0.87) across non-Hispanic white, non-Hispanic African American, Hispanic Latino, and East Asian groups compared to supervised baseline models (sensitivity: 0.39-0.53; AUCPR: 0.3-0.7). SSPUL also exhibited superior fairness as evidenced by the lowest cumulative parity loss. We identified top shared and distinct features among labeled and unlabeled AD patients, including those that are neurological (e.g., memory loss) and non-neurological (e.g., decubitus ulcer). We validated our results using polygenic risk scores, which were higher in labeled and predicted positives than in predicted negatives among non-Hispanic white, Hispanic Latino, and East Asian groups (p < 0.001).},
}
RevDate: 2025-11-27
The evolving etiologies of rapidly progressive dementia: a systematic review.
Translational psychiatry pii:10.1038/s41398-025-03777-7 [Epub ahead of print].
BACKGROUND AND PURPOSE: Rapidly progressive dementia (RPD) represents a heterogeneous group of clinical dementia syndromes characterized by a precipitous decline in cognitive function, typically occurring over weeks to months, with diverse underlying etiologies. To elucidate the evolving etiological spectrum of RPD and investigate temporal trends, we conducted a comprehensive systematic review incorporating temporal subgroup analyses.
METHOD: English-language studies published prior to December 31, 2024 were searched in the Medline (PubMed), ISI Web of Science and the Cochrane Library databases. Study design, study period, patient characteristics, etiologies of RPD in each study were retrieved.
RESULT: We encompassed 13 studies in our final systematic review, involving a total of 1,701 patients with RPD. Our findings revealed that neurodegenerative diseases (459 cases, 27.0%), neuroimmune diseases (327 cases, 19.2%), and central nervous system infections (295 cases, 17.3%) represented the most prevalent etiological categories associated with RPD. Among distinct disease entities, autoimmune encephalitis (AE, 266 cases, 15.6%), Alzheimer's disease (214 cases, 12.6%), and Creutzfeldt-Jakob disease (214 cases, 12.6%) were identified as the three most frequently observed etiologies of RPD. Temporally, neuroimmune diseases, particularly AE, exhibited a significant upward trend in prevalence over the study period.
CONCLUSION: These findings highlight the evolving etiological spectrum of RPD and emphasize the need for ongoing research and targeted diagnostic strategies to address the rising incidence of neuroimmune-related RPD.
Additional Links: PMID-41309536
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309536,
year = {2025},
author = {Lei, MH and Cao, LJ and Liu, R and Zhang, YE and Zhang, SR and Zhuang, LY and Mo, YJ and Cheng, L and Lai, QL and Qiao, S},
title = {The evolving etiologies of rapidly progressive dementia: a systematic review.},
journal = {Translational psychiatry},
volume = {},
number = {},
pages = {},
doi = {10.1038/s41398-025-03777-7},
pmid = {41309536},
issn = {2158-3188},
support = {LQ23H090004//Natural Science Foundation of Zhejiang Province (Zhejiang Provincial Natural Science Foundation)/ ; },
abstract = {BACKGROUND AND PURPOSE: Rapidly progressive dementia (RPD) represents a heterogeneous group of clinical dementia syndromes characterized by a precipitous decline in cognitive function, typically occurring over weeks to months, with diverse underlying etiologies. To elucidate the evolving etiological spectrum of RPD and investigate temporal trends, we conducted a comprehensive systematic review incorporating temporal subgroup analyses.
METHOD: English-language studies published prior to December 31, 2024 were searched in the Medline (PubMed), ISI Web of Science and the Cochrane Library databases. Study design, study period, patient characteristics, etiologies of RPD in each study were retrieved.
RESULT: We encompassed 13 studies in our final systematic review, involving a total of 1,701 patients with RPD. Our findings revealed that neurodegenerative diseases (459 cases, 27.0%), neuroimmune diseases (327 cases, 19.2%), and central nervous system infections (295 cases, 17.3%) represented the most prevalent etiological categories associated with RPD. Among distinct disease entities, autoimmune encephalitis (AE, 266 cases, 15.6%), Alzheimer's disease (214 cases, 12.6%), and Creutzfeldt-Jakob disease (214 cases, 12.6%) were identified as the three most frequently observed etiologies of RPD. Temporally, neuroimmune diseases, particularly AE, exhibited a significant upward trend in prevalence over the study period.
CONCLUSION: These findings highlight the evolving etiological spectrum of RPD and emphasize the need for ongoing research and targeted diagnostic strategies to address the rising incidence of neuroimmune-related RPD.},
}
RevDate: 2025-11-27
SpecQuality: A Tool for Reliable Spectral Quality Assessment in Proteomics and Proteogenomics.
Journal of the American Society for Mass Spectrometry [Epub ahead of print].
Proteogenomics integrates genomics and mass spectrometry (MS) data to understand complex biological systems, disease mechanisms, and potential biomarkers. However, the high volume and noise in MS data present computational and interpretational challenges in proteogenomic studies where, despite best efforts, many spectra are often left unassigned. We developed SpecQuality, an easy-to-use tool for MS/MS quality assessment. We evaluated ten spectral features and, using the top features, developed a machine learning-based model to predict the quality of MS/MS spectra through a spectral quality score (SQS). SpecQuality can be used prior to database search or for postsearch validation of peptide spectrum matches (PSMs). This enables rapid prioritization of high-quality PSMs from proteomics and proteogenomics searches, thereby reducing false-positive identifications. We demonstrated its utility in proteogenomics applications by evaluating its performance on two data sets with ∼2.7 million spectra from Alzheimer's disease, where it successfully highlighted high quality spectra. The spectra that matched novel, variant, and modified peptides in the proteogenomics search were observed to be of high spectral quality. Additionally, a direct comparison with manually curated variant identifications demonstrated its capacity to mitigate false positives and enhance reliability. SpecQuality is an open-source, freely available, easy-to-use, simple, and versatile tool developed in both Python and Perl that can be leveraged in many proteomics pipelines. It can be easily used as a standalone tool or integrated as a part of a bioinformatics data analysis pipeline. SpecQuality provides a scalable and accessible approach to spectral prioritization, advancing data integrity in proteomics and proteogenomics. SpecQuality is available at https://github.com/alkayadav10/SpecQuality.
Additional Links: PMID-41309440
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309440,
year = {2025},
author = {Yadav, A and Dhawan, U},
title = {SpecQuality: A Tool for Reliable Spectral Quality Assessment in Proteomics and Proteogenomics.},
journal = {Journal of the American Society for Mass Spectrometry},
volume = {},
number = {},
pages = {},
doi = {10.1021/jasms.5c00168},
pmid = {41309440},
issn = {1879-1123},
abstract = {Proteogenomics integrates genomics and mass spectrometry (MS) data to understand complex biological systems, disease mechanisms, and potential biomarkers. However, the high volume and noise in MS data present computational and interpretational challenges in proteogenomic studies where, despite best efforts, many spectra are often left unassigned. We developed SpecQuality, an easy-to-use tool for MS/MS quality assessment. We evaluated ten spectral features and, using the top features, developed a machine learning-based model to predict the quality of MS/MS spectra through a spectral quality score (SQS). SpecQuality can be used prior to database search or for postsearch validation of peptide spectrum matches (PSMs). This enables rapid prioritization of high-quality PSMs from proteomics and proteogenomics searches, thereby reducing false-positive identifications. We demonstrated its utility in proteogenomics applications by evaluating its performance on two data sets with ∼2.7 million spectra from Alzheimer's disease, where it successfully highlighted high quality spectra. The spectra that matched novel, variant, and modified peptides in the proteogenomics search were observed to be of high spectral quality. Additionally, a direct comparison with manually curated variant identifications demonstrated its capacity to mitigate false positives and enhance reliability. SpecQuality is an open-source, freely available, easy-to-use, simple, and versatile tool developed in both Python and Perl that can be leveraged in many proteomics pipelines. It can be easily used as a standalone tool or integrated as a part of a bioinformatics data analysis pipeline. SpecQuality provides a scalable and accessible approach to spectral prioritization, advancing data integrity in proteomics and proteogenomics. SpecQuality is available at https://github.com/alkayadav10/SpecQuality.},
}
RevDate: 2025-11-27
Sirtuins in Alzheimer's disease: mechanistic insights and therapeutic opportunities.
Trends in pharmacological sciences pii:S0165-6147(25)00240-8 [Epub ahead of print].
Alzheimer's disease (AD) is an irreversible neurodegenerative disorder characterized by progressive cognitive decline and complex neuropathology. Its main features include amyloid-β (Aβ) plaques, tau neurofibrillary tangles (NFTs), and neuroinflammation. Current therapies provide only limited symptomatic relief and cannot stop disease progression, highlighting the urgent need for disease-modifying strategies. Recent research has revealed multiple roles of sirtuins in AD pathology, positioning them as promising therapeutic targets. Studies using small-molecule compounds to target sirtuins, in both cellular and animal models and clinical analyses of AD patients, demonstrate their therapeutic potential. This review discusses the distinct roles of individual sirtuin isoforms in AD pathogenesis and evaluates the therapeutic evidence for small-molecule sirtuin modulators.
Additional Links: PMID-41309432
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309432,
year = {2025},
author = {Bai, N and Liu, S and Wei, J and Zheng, B and Wang, W and Li, X and Yang, J and Song, X and Wang, L and Yi, F and Cao, L},
title = {Sirtuins in Alzheimer's disease: mechanistic insights and therapeutic opportunities.},
journal = {Trends in pharmacological sciences},
volume = {},
number = {},
pages = {},
doi = {10.1016/j.tips.2025.10.016},
pmid = {41309432},
issn = {1873-3735},
abstract = {Alzheimer's disease (AD) is an irreversible neurodegenerative disorder characterized by progressive cognitive decline and complex neuropathology. Its main features include amyloid-β (Aβ) plaques, tau neurofibrillary tangles (NFTs), and neuroinflammation. Current therapies provide only limited symptomatic relief and cannot stop disease progression, highlighting the urgent need for disease-modifying strategies. Recent research has revealed multiple roles of sirtuins in AD pathology, positioning them as promising therapeutic targets. Studies using small-molecule compounds to target sirtuins, in both cellular and animal models and clinical analyses of AD patients, demonstrate their therapeutic potential. This review discusses the distinct roles of individual sirtuin isoforms in AD pathogenesis and evaluates the therapeutic evidence for small-molecule sirtuin modulators.},
}
RevDate: 2025-11-27
Corrigendum to "Aging as an active player in Alzheimer's disease classification: Insights from feature selection in BrainAge models" [NeuroImage (2025): 121548].
Additional Links: PMID-41309397
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309397,
year = {2025},
author = {Garcia Condado, J and Verdugo Recuero, I and Elorriaga, IT and Birkenbihl, C and Carrigan, M and Diez, I and Buckley, RF and Erramuzpe, A and Cortes, JM and , },
title = {Corrigendum to "Aging as an active player in Alzheimer's disease classification: Insights from feature selection in BrainAge models" [NeuroImage (2025): 121548].},
journal = {NeuroImage},
volume = {},
number = {},
pages = {121604},
doi = {10.1016/j.neuroimage.2025.121604},
pmid = {41309397},
issn = {1095-9572},
}
RevDate: 2025-11-27
Comparative effectiveness of SGLT2 sodium-glucose cotransporter-2 inhibitors and GLP-1 glucagon-like peptide-1 receptor agonists for incident dementia: A retrospective multicohort study.
Diabetes, obesity & metabolism [Epub ahead of print].
OBJECTIVE: This study provides real-world evidence on the comparative effectiveness of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and sodium-glucose cotransporter-2 inhibitors (SGLT2i) regarding the risk of incident dementia in adults with type 2 diabetes mellitus (T2DM).
RESEARCH DESIGN AND METHODS: This cohort study utilised electronic health records from TriNetX (1 April 2013 to 31 December 2019). Adults with T2DM on metformin who initiated GLP-1 RAs or SGLT2i were analysed using Cox proportional hazards models in 1:1 propensity score-matched cohorts. The primary outcome was all-cause dementia; secondary outcomes included vascular and Alzheimer's dementia. Confounding variables were adjusted.
RESULTS: Among 16 271 participants using GLP-1 RAs and 16 271 using SGLT2i over a median 6.3-year follow-up, 581 GLP-1 RA users and 572 SGLT2i users developed dementia. No significant differences were observed in all-cause dementia (hazard ratio: 1.01, 95% confidence interval: 0.90-1.13), vascular, or Alzheimer's dementia. Subgroup analyses confirmed these findings.
CONCLUSIONS: SGLT2i use was not associated with increased risk of dementia compared with GLP-1RAs in patients with T2DM. Further research is warranted to explore the effects of (newer) incretin-based (co-)agonists on cognitive outcomes.
Additional Links: PMID-41309383
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309383,
year = {2025},
author = {Anagnostakis, F and Kokkorakis, M and Nagarajan, S and Anastasiou, G and Mantzoros, CS},
title = {Comparative effectiveness of SGLT2 sodium-glucose cotransporter-2 inhibitors and GLP-1 glucagon-like peptide-1 receptor agonists for incident dementia: A retrospective multicohort study.},
journal = {Diabetes, obesity & metabolism},
volume = {},
number = {},
pages = {},
doi = {10.1111/dom.70336},
pmid = {41309383},
issn = {1463-1326},
abstract = {OBJECTIVE: This study provides real-world evidence on the comparative effectiveness of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and sodium-glucose cotransporter-2 inhibitors (SGLT2i) regarding the risk of incident dementia in adults with type 2 diabetes mellitus (T2DM).
RESEARCH DESIGN AND METHODS: This cohort study utilised electronic health records from TriNetX (1 April 2013 to 31 December 2019). Adults with T2DM on metformin who initiated GLP-1 RAs or SGLT2i were analysed using Cox proportional hazards models in 1:1 propensity score-matched cohorts. The primary outcome was all-cause dementia; secondary outcomes included vascular and Alzheimer's dementia. Confounding variables were adjusted.
RESULTS: Among 16 271 participants using GLP-1 RAs and 16 271 using SGLT2i over a median 6.3-year follow-up, 581 GLP-1 RA users and 572 SGLT2i users developed dementia. No significant differences were observed in all-cause dementia (hazard ratio: 1.01, 95% confidence interval: 0.90-1.13), vascular, or Alzheimer's dementia. Subgroup analyses confirmed these findings.
CONCLUSIONS: SGLT2i use was not associated with increased risk of dementia compared with GLP-1RAs in patients with T2DM. Further research is warranted to explore the effects of (newer) incretin-based (co-)agonists on cognitive outcomes.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
The calpain-mediated proteolytic cleavage peptide P10 modulates Tau phosphorylation in Alzheimer's disease.
Advances in protein chemistry and structural biology, 148:481-505.
Alzheimer's disease is a prominent neurological disorder, which leads to progressive dementia. The microtubule-associated protein Tau is considered one of the major causes of Alzheimer's disease. Hyper-phosphorylation of Tau is considered to be closely associated with the generation of Tau pathology. CDK5 is one of the prominent neuronal kinases, under normal conditions, the activity of CDK5 is regulated by p35 protein. The stress conditions result in calpain-mediated proteolytic cleavage of p35 leading to the generation of p25 and p10. CDK5/p25 complex is reported to have a relatively more half-life which causes hyperphosphorylation of many proteins leading to neurotoxicity but the role of p10 is still needed to be explored. In the present review, we hypothesized the role of p10 as CDK5/p25 inhibitor. The current research has demonstrated that p10 provides survival signals to cells. In the current scenario, several CDK5 inhibitors commonly have a drawback of non-specificity. Here based on complied studies we hypothesize that the p10 could have the potency to inhibit the activity of CDK5, which ultimately downregulate the hyperphosphorylation of Tau. Thus reducing the levels of phospho-Tau p10 could emerge as a novel therapeutic peptide against Alzheimer's disease. The proposed hypothesis would open new gates for research in the field of Alzheimer's and further would bring a new ray of hope for the disease.
Additional Links: PMID-41309199
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309199,
year = {2025},
author = {Chinnathambi, S and Dubey, T and Rangappa, N},
title = {The calpain-mediated proteolytic cleavage peptide P10 modulates Tau phosphorylation in Alzheimer's disease.},
journal = {Advances in protein chemistry and structural biology},
volume = {148},
number = {},
pages = {481-505},
doi = {10.1016/bs.apcsb.2025.08.009},
pmid = {41309199},
issn = {1876-1631},
mesh = {*Alzheimer Disease/metabolism/pathology/drug therapy ; Humans ; *tau Proteins/metabolism ; Phosphorylation/drug effects ; *Calpain/metabolism ; Proteolysis ; Cyclin-Dependent Kinase 5/metabolism/antagonists & inhibitors ; Animals ; },
abstract = {Alzheimer's disease is a prominent neurological disorder, which leads to progressive dementia. The microtubule-associated protein Tau is considered one of the major causes of Alzheimer's disease. Hyper-phosphorylation of Tau is considered to be closely associated with the generation of Tau pathology. CDK5 is one of the prominent neuronal kinases, under normal conditions, the activity of CDK5 is regulated by p35 protein. The stress conditions result in calpain-mediated proteolytic cleavage of p35 leading to the generation of p25 and p10. CDK5/p25 complex is reported to have a relatively more half-life which causes hyperphosphorylation of many proteins leading to neurotoxicity but the role of p10 is still needed to be explored. In the present review, we hypothesized the role of p10 as CDK5/p25 inhibitor. The current research has demonstrated that p10 provides survival signals to cells. In the current scenario, several CDK5 inhibitors commonly have a drawback of non-specificity. Here based on complied studies we hypothesize that the p10 could have the potency to inhibit the activity of CDK5, which ultimately downregulate the hyperphosphorylation of Tau. Thus reducing the levels of phospho-Tau p10 could emerge as a novel therapeutic peptide against Alzheimer's disease. The proposed hypothesis would open new gates for research in the field of Alzheimer's and further would bring a new ray of hope for the disease.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/metabolism/pathology/drug therapy
Humans
*tau Proteins/metabolism
Phosphorylation/drug effects
*Calpain/metabolism
Proteolysis
Cyclin-Dependent Kinase 5/metabolism/antagonists & inhibitors
Animals
RevDate: 2025-11-27
CmpDate: 2025-11-27
Chaperone machinery in neurodegeneration: A spotlight on protein misfolding diseases.
Advances in protein chemistry and structural biology, 148:455-480.
Proteins misfolding in neurodegenerative disorders pose a significant challenge to human health and this necessitates a deeper understanding of the fundamental molecular mechanisms. Molecular chaperones are a diverse group of specialized proteins, which are extensively involved in maintaining cellular protein homeostasis and thus preventing aggregation of misfolded proteins. Pathological advancement in several neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD) is characterized by the rampant accretion of misfolded proteins due to chaperonic failure, leading to progressive neuronal dysfunctioning and eventually cell death. Such as in AD, Hsp70 and Hsp90 chaperones are known to interact with β-amyloid and tau proteins, thus preventing their subsequent aggregation with concomitant refolding into native conformations. In PD, chaperones are involved in assisting mitigation of α-Syn misfolding and aggregation, thereby maintaining the normal neuronal functions and their viability. Similarly in HD, chaperones modulate aberrant misfolding of huntingtin protein and its aggregation, thus highlighting prospective therapeutic targets for disease intervention. Nevertheless, further investigating and understanding the explicit roles of chaperones in modulating several protein misfolding diseases holds potential for the development of novel therapeutic approaches. Moreover, targeting such specialized chaperone machinery in restoring protein homeostasis and alleviating subsequent protein aggregation could be considered as a promising approach in managing neurodegenerative disorders.
Additional Links: PMID-41309198
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309198,
year = {2025},
author = {Sood, A and Dey, M and Tyagi, A and Sharma, DK and Mehrotra, A},
title = {Chaperone machinery in neurodegeneration: A spotlight on protein misfolding diseases.},
journal = {Advances in protein chemistry and structural biology},
volume = {148},
number = {},
pages = {455-480},
doi = {10.1016/bs.apcsb.2025.08.007},
pmid = {41309198},
issn = {1876-1631},
mesh = {Humans ; *Proteostasis Deficiencies/metabolism/pathology ; *Neurodegenerative Diseases/metabolism/pathology ; *Molecular Chaperones/metabolism ; Protein Folding ; Animals ; },
abstract = {Proteins misfolding in neurodegenerative disorders pose a significant challenge to human health and this necessitates a deeper understanding of the fundamental molecular mechanisms. Molecular chaperones are a diverse group of specialized proteins, which are extensively involved in maintaining cellular protein homeostasis and thus preventing aggregation of misfolded proteins. Pathological advancement in several neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD) is characterized by the rampant accretion of misfolded proteins due to chaperonic failure, leading to progressive neuronal dysfunctioning and eventually cell death. Such as in AD, Hsp70 and Hsp90 chaperones are known to interact with β-amyloid and tau proteins, thus preventing their subsequent aggregation with concomitant refolding into native conformations. In PD, chaperones are involved in assisting mitigation of α-Syn misfolding and aggregation, thereby maintaining the normal neuronal functions and their viability. Similarly in HD, chaperones modulate aberrant misfolding of huntingtin protein and its aggregation, thus highlighting prospective therapeutic targets for disease intervention. Nevertheless, further investigating and understanding the explicit roles of chaperones in modulating several protein misfolding diseases holds potential for the development of novel therapeutic approaches. Moreover, targeting such specialized chaperone machinery in restoring protein homeostasis and alleviating subsequent protein aggregation could be considered as a promising approach in managing neurodegenerative disorders.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Proteostasis Deficiencies/metabolism/pathology
*Neurodegenerative Diseases/metabolism/pathology
*Molecular Chaperones/metabolism
Protein Folding
Animals
RevDate: 2025-11-27
CmpDate: 2025-11-27
Protein misfolding and its dual role in neurodegeneration and cancer progression.
Advances in protein chemistry and structural biology, 148:355-377.
Protein misfolding is a fundamental biological process with profound implications for human health and disease. Typically, proteins assume precise three-dimensional structures to perform their functions, a process safeguarded by the proteostasis network, which comprises molecular chaperones, the ubiquitin-proteasome system (UPS), and autophagy. However, genetic mutations, oxidative stress, and environmental insults can disrupt folding, leading to the accumulation of non-functional or toxic conformations. In neurodegenerative diseases such as Huntington's disease (HD), Parkinson's disease (PD), Alzheimer's disease (AD), Amyotrophic lateral Sclerosis (ALS), chronic misfolding results in toxic protein aggregates like amyloid-β, tau, and α-synuclein. These disrupt synaptic function, induce oxidative and nitrosative stress, and trigger apoptosis, ultimately leading to progressive neuronal loss. Dysregulation of the unfolded protein response (UPR) and weakened proteostasis with aging exacerbate disease pathology. In contrast, cancer cells utilize protein misfolding to enhance their survival and progression. Misfolded oncoproteins, such as mutant p53, not only evade degradation but also acquire oncogenic properties. Tumor cells hijack the UPR and chaperone networks, upregulate heat shock proteins, and manipulate oxidative stress responses to withstand hypoxia, nutrient deprivation, and rapid proliferation. Cancer stem cells (CSCs) further adapt to proteotoxic stress, contributing to tumor heterogeneity, therapy resistance, and immune evasion. The dual role of protein misfolding, driving degeneration in neurons while supporting proliferation in tumors, underscores its centrality in disease biology. Future research should focus on identifying early biomarkers of proteostasis imbalance and exploiting shared molecular pathways for the development of novel therapeutic interventions.
Additional Links: PMID-41309196
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309196,
year = {2025},
author = {Jamwal, RS and Sharma, B and Minerva, and Gupta, A and Misri, S and Shankaryan, R and Shah, R and Kumar, R},
title = {Protein misfolding and its dual role in neurodegeneration and cancer progression.},
journal = {Advances in protein chemistry and structural biology},
volume = {148},
number = {},
pages = {355-377},
doi = {10.1016/bs.apcsb.2025.10.001},
pmid = {41309196},
issn = {1876-1631},
mesh = {Humans ; *Neoplasms/metabolism/pathology ; *Protein Folding ; *Neurodegenerative Diseases/metabolism/pathology ; Animals ; Disease Progression ; Unfolded Protein Response ; },
abstract = {Protein misfolding is a fundamental biological process with profound implications for human health and disease. Typically, proteins assume precise three-dimensional structures to perform their functions, a process safeguarded by the proteostasis network, which comprises molecular chaperones, the ubiquitin-proteasome system (UPS), and autophagy. However, genetic mutations, oxidative stress, and environmental insults can disrupt folding, leading to the accumulation of non-functional or toxic conformations. In neurodegenerative diseases such as Huntington's disease (HD), Parkinson's disease (PD), Alzheimer's disease (AD), Amyotrophic lateral Sclerosis (ALS), chronic misfolding results in toxic protein aggregates like amyloid-β, tau, and α-synuclein. These disrupt synaptic function, induce oxidative and nitrosative stress, and trigger apoptosis, ultimately leading to progressive neuronal loss. Dysregulation of the unfolded protein response (UPR) and weakened proteostasis with aging exacerbate disease pathology. In contrast, cancer cells utilize protein misfolding to enhance their survival and progression. Misfolded oncoproteins, such as mutant p53, not only evade degradation but also acquire oncogenic properties. Tumor cells hijack the UPR and chaperone networks, upregulate heat shock proteins, and manipulate oxidative stress responses to withstand hypoxia, nutrient deprivation, and rapid proliferation. Cancer stem cells (CSCs) further adapt to proteotoxic stress, contributing to tumor heterogeneity, therapy resistance, and immune evasion. The dual role of protein misfolding, driving degeneration in neurons while supporting proliferation in tumors, underscores its centrality in disease biology. Future research should focus on identifying early biomarkers of proteostasis imbalance and exploiting shared molecular pathways for the development of novel therapeutic interventions.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Neoplasms/metabolism/pathology
*Protein Folding
*Neurodegenerative Diseases/metabolism/pathology
Animals
Disease Progression
Unfolded Protein Response
RevDate: 2025-11-27
CmpDate: 2025-11-27
Proteostasis and pathogenesis: Unraveling the complexity of protein misfolding disorders.
Advances in protein chemistry and structural biology, 148:299-353.
Within the cellular milieu, protein molecules must fold into precise three-dimensional structures to attain functionality. Protein chains can assume many misfolded states during this critical process. Such errant configurations are unstable and can aggregate into toxic misfolded conformations. In protein misfolding disorders, polypeptides are folded in an aberrant manner, resulting in non-functional and often pathogenic states. Protein folding is fundamental to biological function, and disruption of the process can result in toxic aggregates, such as oligomers and amyloid fibrils, which are implicated in a variety of diseases, particularly neurodegenerative diseases such as Alzheimer's and Parkinson's. Here, we examine the delicate interplay of forces that determine the native conformation of proteins and how perturbations in this balance lead to disease. A critical aspect of our discussion is the cell's proteostasis network, a complex network of molecular chaperones and regulators responsible for regulating protein folding and maintaining the health of the cell. In this chapter, we discuss how intrinsic protein properties, post-translational modifications, and extrinsic environmental factors can destabilize proteins, thereby resulting in their misfolding. Several pathogenic mechanisms will be elucidated, including the progression from a misfolded protein to the development of disease phenotypes. Next, the chapter will present an overview of the current therapeutic approaches to mitigate the diseases caused by protein misfolding. Using the latest findings in clinical and experimental research, we will evaluate the therapeutic landscape, ranging from small-molecule inhibitors to chaperone-based therapies.
Additional Links: PMID-41309195
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309195,
year = {2025},
author = {Nair, T and Sarkar, D and Murmu, S and Singh Rawat, R and Singha, B},
title = {Proteostasis and pathogenesis: Unraveling the complexity of protein misfolding disorders.},
journal = {Advances in protein chemistry and structural biology},
volume = {148},
number = {},
pages = {299-353},
doi = {10.1016/bs.apcsb.2025.08.013},
pmid = {41309195},
issn = {1876-1631},
mesh = {Humans ; *Proteostasis Deficiencies/metabolism/pathology ; *Proteostasis ; Protein Folding ; Animals ; Protein Processing, Post-Translational ; Molecular Chaperones/metabolism ; *Neurodegenerative Diseases/metabolism/pathology ; },
abstract = {Within the cellular milieu, protein molecules must fold into precise three-dimensional structures to attain functionality. Protein chains can assume many misfolded states during this critical process. Such errant configurations are unstable and can aggregate into toxic misfolded conformations. In protein misfolding disorders, polypeptides are folded in an aberrant manner, resulting in non-functional and often pathogenic states. Protein folding is fundamental to biological function, and disruption of the process can result in toxic aggregates, such as oligomers and amyloid fibrils, which are implicated in a variety of diseases, particularly neurodegenerative diseases such as Alzheimer's and Parkinson's. Here, we examine the delicate interplay of forces that determine the native conformation of proteins and how perturbations in this balance lead to disease. A critical aspect of our discussion is the cell's proteostasis network, a complex network of molecular chaperones and regulators responsible for regulating protein folding and maintaining the health of the cell. In this chapter, we discuss how intrinsic protein properties, post-translational modifications, and extrinsic environmental factors can destabilize proteins, thereby resulting in their misfolding. Several pathogenic mechanisms will be elucidated, including the progression from a misfolded protein to the development of disease phenotypes. Next, the chapter will present an overview of the current therapeutic approaches to mitigate the diseases caused by protein misfolding. Using the latest findings in clinical and experimental research, we will evaluate the therapeutic landscape, ranging from small-molecule inhibitors to chaperone-based therapies.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Proteostasis Deficiencies/metabolism/pathology
*Proteostasis
Protein Folding
Animals
Protein Processing, Post-Translational
Molecular Chaperones/metabolism
*Neurodegenerative Diseases/metabolism/pathology
RevDate: 2025-11-27
CmpDate: 2025-11-27
Unraveling tau's fold: Structural dynamics in Alzheimer's pathogenesis.
Advances in protein chemistry and structural biology, 148:29-56.
Alzheimer's disease (AD), among the diseases associated with dementia, is the most prevalent. It has been estimated that over 55 million people older than 65 years-old are living with dementia worldwide. Two-thirds of the AD population are women. It is estimated that by 2050 there will be 139 million people with dementia. AD is a neurodegenerative, progressive and irreversible process, affecting the patient's daily life activities. The pathological neurodegenerative process of AD begins 15-20 years before the appearance of the first clinical symptoms. The histopathological analysis reveals the presence of neurofibrillary tangles (NFTs) and neuritic plaques [1] the main hallmarks of AD. In this work, we are describing the NFTs that are made up of paired helical filaments of tau protein, which undergo post-translational modifications such as hyperphosphorylation and truncation, favoring conformational changes of the molecule. The most relevant information about the pathological processing of the tau protein is presented, focusing on the truncation at Glu391 (minimal filament nucleus, PHF-core) as a pathological inducing event of the tau protein and as an early biomarker of AD. Based on reports and our evidence, we suggest that the hyperphosphorylated tau protein participates as the neuroprotective event against this highly toxic PHF-core.
Additional Links: PMID-41309194
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309194,
year = {2025},
author = {Montaño, S and Luna-Viramontes, NI and Cuevas, E and Martínez-Cuevas, PP and Pacheco-Herrero, M and León-López, L and Armenta, AD and Luna-Muñoz, J},
title = {Unraveling tau's fold: Structural dynamics in Alzheimer's pathogenesis.},
journal = {Advances in protein chemistry and structural biology},
volume = {148},
number = {},
pages = {29-56},
doi = {10.1016/bs.apcsb.2025.08.005},
pmid = {41309194},
issn = {1876-1631},
mesh = {*tau Proteins/metabolism/chemistry ; Humans ; *Alzheimer Disease/metabolism/pathology ; Neurofibrillary Tangles/metabolism/pathology ; Phosphorylation ; Protein Processing, Post-Translational ; },
abstract = {Alzheimer's disease (AD), among the diseases associated with dementia, is the most prevalent. It has been estimated that over 55 million people older than 65 years-old are living with dementia worldwide. Two-thirds of the AD population are women. It is estimated that by 2050 there will be 139 million people with dementia. AD is a neurodegenerative, progressive and irreversible process, affecting the patient's daily life activities. The pathological neurodegenerative process of AD begins 15-20 years before the appearance of the first clinical symptoms. The histopathological analysis reveals the presence of neurofibrillary tangles (NFTs) and neuritic plaques [1] the main hallmarks of AD. In this work, we are describing the NFTs that are made up of paired helical filaments of tau protein, which undergo post-translational modifications such as hyperphosphorylation and truncation, favoring conformational changes of the molecule. The most relevant information about the pathological processing of the tau protein is presented, focusing on the truncation at Glu391 (minimal filament nucleus, PHF-core) as a pathological inducing event of the tau protein and as an early biomarker of AD. Based on reports and our evidence, we suggest that the hyperphosphorylated tau protein participates as the neuroprotective event against this highly toxic PHF-core.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*tau Proteins/metabolism/chemistry
Humans
*Alzheimer Disease/metabolism/pathology
Neurofibrillary Tangles/metabolism/pathology
Phosphorylation
Protein Processing, Post-Translational
RevDate: 2025-11-27
CmpDate: 2025-11-27
Protein misfolding and neurodegeneration: Mechanisms, implications, and therapeutic strategies.
Advances in protein chemistry and structural biology, 148:135-177.
Protein misfolding and aggregation play a pivotal role in the development of neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's disease, and other related disorders. Proper protein folding is essential for cellular function, but due to the complexity of the folding process and external factors like genetic mutations, oxidative stress, and aging, misfolding is inevitable. These misfolded proteins often aggregate into toxic forms that disrupt cellular processes, leading to neuronal damage and cognitive decline. This chapter provides a comprehensive overview of molecular mechanisms behind protein misfolding, highlighting how these abnormal structures contribute to neurodegeneration. It also explores the role of the proteostasis network and its therapeutic potential in alleviating these processes. Focusing on multitarget therapeutic strategies, the chapter offers insights into promising approaches for addressing the root causes of neurodegenerative diseases while identifying key research gaps that could shape future treatment developments. By blending current knowledge with emerging therapeutic directions, this chapter provides a comprehensive and engaging perspective on combating the challenges of protein misfolding in neurodegeneration.
Additional Links: PMID-41309190
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41309190,
year = {2025},
author = {Shah, A and Karthikeyan, T and Hashem, S and Kumar, R and Bhat, AA and Macha, MA},
title = {Protein misfolding and neurodegeneration: Mechanisms, implications, and therapeutic strategies.},
journal = {Advances in protein chemistry and structural biology},
volume = {148},
number = {},
pages = {135-177},
doi = {10.1016/bs.apcsb.2025.08.012},
pmid = {41309190},
issn = {1876-1631},
mesh = {Humans ; *Protein Folding ; *Neurodegenerative Diseases/metabolism/pathology/drug therapy/therapy ; *Proteostasis Deficiencies/metabolism/pathology/therapy ; Animals ; Proteostasis ; },
abstract = {Protein misfolding and aggregation play a pivotal role in the development of neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's disease, and other related disorders. Proper protein folding is essential for cellular function, but due to the complexity of the folding process and external factors like genetic mutations, oxidative stress, and aging, misfolding is inevitable. These misfolded proteins often aggregate into toxic forms that disrupt cellular processes, leading to neuronal damage and cognitive decline. This chapter provides a comprehensive overview of molecular mechanisms behind protein misfolding, highlighting how these abnormal structures contribute to neurodegeneration. It also explores the role of the proteostasis network and its therapeutic potential in alleviating these processes. Focusing on multitarget therapeutic strategies, the chapter offers insights into promising approaches for addressing the root causes of neurodegenerative diseases while identifying key research gaps that could shape future treatment developments. By blending current knowledge with emerging therapeutic directions, this chapter provides a comprehensive and engaging perspective on combating the challenges of protein misfolding in neurodegeneration.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Protein Folding
*Neurodegenerative Diseases/metabolism/pathology/drug therapy/therapy
*Proteostasis Deficiencies/metabolism/pathology/therapy
Animals
Proteostasis
RevDate: 2025-11-27
Development of a VHH that inhibits the binding of neuronal pentraxin 2 to a post-synaptic glutamate receptor, AMPAR.
The Journal of biological chemistry pii:S0021-9258(25)02827-3 [Epub ahead of print].
Neurons connect to each other via synapses to form neural circuits. Recent research has shown that neuropsychiatric disorders and neurological disorders such as autism spectrum disorders and Alzheimer's disease (AD) are synaptic diseases caused by abnormality of synapses. Synaptic organizers are molecules responsible for synapse formation. Neuronal pentraxin 2 (NP2) is synaptic organizer and a secreted protein that is expressed mainly in the hippocampus and cerebellum, and it contributes to synaptic plasticity. NP2 forms clusters with its family proteins NP1 and NPR and binds to postsynaptic amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type receptors (AMPARs). In recent years, research has revealed the disease relevance of NP2. For example, it can be a biomarker of AD, and its overexpression in the peripheral nervous system has been reported to cause chronic itch. However, the mechanism of NP2 function has not been well described at the molecular level. In this study, we developed a variable domain of heavy chain of heavy chain antibody (VHH) against NP2 to elucidate its molecular mechanism of action and to regulate its function of NP2. The obtained VHH N1 showed high specificity and affinity to NP2, and its binding mechanism was elucidated by X-ray crystallography. Furthermore, VHH N1 inhibited the binding of NP2 to AMPARs, and this inhibitory activity was confirmed in cells. These results provide useful insights into the molecular mechanism of NP2 function and highlight the potential application of VHH N1 as a detection agent for NP2 or as a therapeutic agent for chronic itch.
Additional Links: PMID-41308985
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308985,
year = {2025},
author = {Yokoo, T and Nakakido, M and Matsuda, K and Caaveiro, JMM and Fernandez-Perez, J and Yuzaki, M and Tsumoto, K},
title = {Development of a VHH that inhibits the binding of neuronal pentraxin 2 to a post-synaptic glutamate receptor, AMPAR.},
journal = {The Journal of biological chemistry},
volume = {},
number = {},
pages = {110975},
doi = {10.1016/j.jbc.2025.110975},
pmid = {41308985},
issn = {1083-351X},
abstract = {Neurons connect to each other via synapses to form neural circuits. Recent research has shown that neuropsychiatric disorders and neurological disorders such as autism spectrum disorders and Alzheimer's disease (AD) are synaptic diseases caused by abnormality of synapses. Synaptic organizers are molecules responsible for synapse formation. Neuronal pentraxin 2 (NP2) is synaptic organizer and a secreted protein that is expressed mainly in the hippocampus and cerebellum, and it contributes to synaptic plasticity. NP2 forms clusters with its family proteins NP1 and NPR and binds to postsynaptic amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type receptors (AMPARs). In recent years, research has revealed the disease relevance of NP2. For example, it can be a biomarker of AD, and its overexpression in the peripheral nervous system has been reported to cause chronic itch. However, the mechanism of NP2 function has not been well described at the molecular level. In this study, we developed a variable domain of heavy chain of heavy chain antibody (VHH) against NP2 to elucidate its molecular mechanism of action and to regulate its function of NP2. The obtained VHH N1 showed high specificity and affinity to NP2, and its binding mechanism was elucidated by X-ray crystallography. Furthermore, VHH N1 inhibited the binding of NP2 to AMPARs, and this inhibitory activity was confirmed in cells. These results provide useful insights into the molecular mechanism of NP2 function and highlight the potential application of VHH N1 as a detection agent for NP2 or as a therapeutic agent for chronic itch.},
}
RevDate: 2025-11-27
Neural stem cells improve the function of learning and memory in the ibotenic acid-induced Alzheimer's disease mice.
Behavioural brain research pii:S0166-4328(25)00552-2 [Epub ahead of print].
Neural stem cells (NSCs) showed a promising approach to treat Alzheimer's disease (AD). This study aims to investigate whether mouse NSCs transplantation can improve the function of learning and memory of AD mouse model and the underlying mechanism. NSCs was stereotaxically injected into the hippocampi of ibotenic acid (IBO)-induced AD mice. Behavioral tests were conducted to evaluate neurological function. Golgi and immunofluorescence staining were used to assess the injury of the nerves. Additionally, enhanced green fluorescent protein (EGFP) labeling was employed to evaluate the differentiation of NSCs. Compared with model group, NSCs transplantation shortened the escape latency and increased the crossing platform quadrant time, number of crossing the platform (p < 0.01). The preference index and active avoidance rate was increased after NSCs transplantation when compared with the model group (p < 0.05 or p < 0.01). NSCs augmented not only the number of neurons in the hippocampal CA1 region, but also the dendritic spine density, dendritic complexity and ACh content in the hippocampus (p < 0.05 or p < 0.01). Furthermore, NSCs also elevated the levels of BDNF in the hippocampus of AD mice (p < 0.01). It also showed that a portion of transplanted NSCs differentiated into neurons at four weeks post-transplantation. These results demonstrated that NSCs improved learning and memory function in the AD model by ameliorating neuron injury and differentiating into neurons.
Additional Links: PMID-41308879
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308879,
year = {2025},
author = {Jia, X and Li, M and Wang, Y and Li, H and Zhang, X and Chen, H and Tian, J and Zhang, G and Wang, T},
title = {Neural stem cells improve the function of learning and memory in the ibotenic acid-induced Alzheimer's disease mice.},
journal = {Behavioural brain research},
volume = {},
number = {},
pages = {115965},
doi = {10.1016/j.bbr.2025.115965},
pmid = {41308879},
issn = {1872-7549},
abstract = {Neural stem cells (NSCs) showed a promising approach to treat Alzheimer's disease (AD). This study aims to investigate whether mouse NSCs transplantation can improve the function of learning and memory of AD mouse model and the underlying mechanism. NSCs was stereotaxically injected into the hippocampi of ibotenic acid (IBO)-induced AD mice. Behavioral tests were conducted to evaluate neurological function. Golgi and immunofluorescence staining were used to assess the injury of the nerves. Additionally, enhanced green fluorescent protein (EGFP) labeling was employed to evaluate the differentiation of NSCs. Compared with model group, NSCs transplantation shortened the escape latency and increased the crossing platform quadrant time, number of crossing the platform (p < 0.01). The preference index and active avoidance rate was increased after NSCs transplantation when compared with the model group (p < 0.05 or p < 0.01). NSCs augmented not only the number of neurons in the hippocampal CA1 region, but also the dendritic spine density, dendritic complexity and ACh content in the hippocampus (p < 0.05 or p < 0.01). Furthermore, NSCs also elevated the levels of BDNF in the hippocampus of AD mice (p < 0.01). It also showed that a portion of transplanted NSCs differentiated into neurons at four weeks post-transplantation. These results demonstrated that NSCs improved learning and memory function in the AD model by ameliorating neuron injury and differentiating into neurons.},
}
RevDate: 2025-11-27
Therapeutic Potential of Third Molar-Derived Dental Pulp Stem Cells in Alzheimer's disease: Current Evidence and Future Directions.
Ageing research reviews pii:S1568-1637(25)00295-8 [Epub ahead of print].
Alzheimer's disease (AD), the most common cause of dementia, is characterized by amyloid-β deposition, tau hyperphosphorylation, neuroinflammation, and progressive neuronal loss, with no curative therapy currently available. Dental pulp stem cells (DPSCs) derived from third molars represent an ethically accessible, minimally invasive, neural crest-derived mesenchymal stem cell source with self-renewal and multi-lineage differentiation potential. Preclinical evidence suggests that DPSCs exert neuroprotective effects by reducing oxidative stress and apoptosis, enhancing neurogenesis through synaptic repair and neuronal differentiation, and modulating neuroinflammation via microglial regulation. Their secretion of neurotrophic factors, including BDNF, GDNF, NGF, NT-3, CNTF, and HGF, further supports neuronal survival and functional recovery in Alzheimer's disease models. Advances in 3D neurosphere and brain organoid models demonstrate the integration of DPSCs into neural circuitry, highlighting their translational potential. Nevertheless, challenges such as cell survival, migration, and functional integration within the diseased brain remain. Importantly, the use of extracted third molars avoids major ethical concerns associated with embryonic stem cells, making DPSCs a clinically relevant candidate for regenerative approaches. Future directions emphasize the development of exosome-based therapies, bioengineered scaffolds, gene-modified DPSC strategies, and personalized autologous interventions. Collectively, current evidence positions third molar-derived DPSCs as a promising avenue for disease-modifying and regenerative therapies in Alzheimer's disease.
Additional Links: PMID-41308841
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308841,
year = {2025},
author = {Wagaskar, P and Suryawanshi, M and Patil, A and Pardeshi, V},
title = {Therapeutic Potential of Third Molar-Derived Dental Pulp Stem Cells in Alzheimer's disease: Current Evidence and Future Directions.},
journal = {Ageing research reviews},
volume = {},
number = {},
pages = {102949},
doi = {10.1016/j.arr.2025.102949},
pmid = {41308841},
issn = {1872-9649},
abstract = {Alzheimer's disease (AD), the most common cause of dementia, is characterized by amyloid-β deposition, tau hyperphosphorylation, neuroinflammation, and progressive neuronal loss, with no curative therapy currently available. Dental pulp stem cells (DPSCs) derived from third molars represent an ethically accessible, minimally invasive, neural crest-derived mesenchymal stem cell source with self-renewal and multi-lineage differentiation potential. Preclinical evidence suggests that DPSCs exert neuroprotective effects by reducing oxidative stress and apoptosis, enhancing neurogenesis through synaptic repair and neuronal differentiation, and modulating neuroinflammation via microglial regulation. Their secretion of neurotrophic factors, including BDNF, GDNF, NGF, NT-3, CNTF, and HGF, further supports neuronal survival and functional recovery in Alzheimer's disease models. Advances in 3D neurosphere and brain organoid models demonstrate the integration of DPSCs into neural circuitry, highlighting their translational potential. Nevertheless, challenges such as cell survival, migration, and functional integration within the diseased brain remain. Importantly, the use of extracted third molars avoids major ethical concerns associated with embryonic stem cells, making DPSCs a clinically relevant candidate for regenerative approaches. Future directions emphasize the development of exosome-based therapies, bioengineered scaffolds, gene-modified DPSC strategies, and personalized autologous interventions. Collectively, current evidence positions third molar-derived DPSCs as a promising avenue for disease-modifying and regenerative therapies in Alzheimer's disease.},
}
RevDate: 2025-11-27
Serotonin-mediated regulation of mitophagy in Alzheimer's disease: mechanistic insights and therapeutic potential.
Ageing research reviews pii:S1568-1637(25)00303-4 [Epub ahead of print].
This review delves into the intricate relationship between serotonin signaling, mitophagy and mitochondrial dysfunction in Alzheimer's disease (AD), with a focus on the mechanistic pathways that link these processes and their potential therapeutic implications. A neurodegenerative condition called Alzheimer's disease is marked by cognitive deterioration. It is increasingly recognized as being influenced by impaired mitochondrial function and mitophagy, the selective degradation of damaged mitochondria. Serotonin, a neurotransmitter traditionally known for its role in mood regulation, has emerged as a critical modulator of mitochondrial dynamics and quality control through its interaction with key pathways such as the PINK1-Parkin and cAMP/PKA signaling pathways. In AD, alterations in serotonin levels and receptor function are associated with disruptions in mitophagy, leading to the accumulation of dysfunctional mitochondria, increased oxidative stress, and subsequent neuronal damage. This review synthesizes current evidence that links serotonin dysregulation to mitochondrial pathology in AD, exploring how impaired serotonin signaling exacerbates mitochondrial dysfunction and contributes to amyloid-beta (Aβ), phosphorylation of Tau (p-Tau) accumulations, and increased neuroinflammation. Additionally, we assessed the therapeutic potential of serotonin-targeting agents, particularly selective serotonin reuptake inhibitors (SSRIs), in restoring mitophagy, enhancing mitochondrial integrity, and attenuating neurodegeneration. By highlighting existing knowledge gaps and key controversies, this review underscores the promise of serotonin-mitochondria pathways as novel therapeutic targets and advocates for focused investigation into receptor-specific, mitophagy-centered interventions for AD.
Additional Links: PMID-41308838
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308838,
year = {2025},
author = {Sultana, OF and Bandaru, M and Bushra, MA and Reddy, PH and Reddy, AP},
title = {Serotonin-mediated regulation of mitophagy in Alzheimer's disease: mechanistic insights and therapeutic potential.},
journal = {Ageing research reviews},
volume = {},
number = {},
pages = {102957},
doi = {10.1016/j.arr.2025.102957},
pmid = {41308838},
issn = {1872-9649},
abstract = {This review delves into the intricate relationship between serotonin signaling, mitophagy and mitochondrial dysfunction in Alzheimer's disease (AD), with a focus on the mechanistic pathways that link these processes and their potential therapeutic implications. A neurodegenerative condition called Alzheimer's disease is marked by cognitive deterioration. It is increasingly recognized as being influenced by impaired mitochondrial function and mitophagy, the selective degradation of damaged mitochondria. Serotonin, a neurotransmitter traditionally known for its role in mood regulation, has emerged as a critical modulator of mitochondrial dynamics and quality control through its interaction with key pathways such as the PINK1-Parkin and cAMP/PKA signaling pathways. In AD, alterations in serotonin levels and receptor function are associated with disruptions in mitophagy, leading to the accumulation of dysfunctional mitochondria, increased oxidative stress, and subsequent neuronal damage. This review synthesizes current evidence that links serotonin dysregulation to mitochondrial pathology in AD, exploring how impaired serotonin signaling exacerbates mitochondrial dysfunction and contributes to amyloid-beta (Aβ), phosphorylation of Tau (p-Tau) accumulations, and increased neuroinflammation. Additionally, we assessed the therapeutic potential of serotonin-targeting agents, particularly selective serotonin reuptake inhibitors (SSRIs), in restoring mitophagy, enhancing mitochondrial integrity, and attenuating neurodegeneration. By highlighting existing knowledge gaps and key controversies, this review underscores the promise of serotonin-mitochondria pathways as novel therapeutic targets and advocates for focused investigation into receptor-specific, mitophagy-centered interventions for AD.},
}
RevDate: 2025-11-27
Multifunctionality of tissue inhibitor of metalloproteinase-1 in central nervous system diseases.
Brain research bulletin pii:S0361-9230(25)00459-9 [Epub ahead of print].
Central nervous system (CNS) diseases are characterized by high morbidity, long disease courses, and irreversible neurological impairment, often resulting from damage to the neurovascular unit (NVU). Tissue inhibitor of metalloproteinase-1 (TIMP-1), the endogenous inhibitor of matrix metalloproteinase-9 (MMP-9), plays a pivotal role in maintaining extracellular matrix (ECM) homeostasis and regulating NVU integrity. Beyond its canonical MMP-inhibitory function, TIMP-1 exerts a wide spectrum of MMP-independent effects as a multifunctional cytokine that interacts with cell surface receptors such as CD63/β1-integrin and low-density lipoprotein receptor-related protein-1 (LRP-1). Through activation of FAK/PI3K-Akt and MAPK signaling pathways, TIMP-1 modulates astrocyte proliferation, neural stem cell adhesion and migration, endothelial barrier stability, and myelin regeneration. Altered TIMP-1 expression is closely associated with the onset, progression, and prognosis of major CNS disorders, including ischemic stroke, epilepsy, multiple sclerosis, and neurodegenerative diseases such as Alzheimer's and Parkinson's disease. Accumulating evidence highlights its dual, context-dependent roles-protective in acute neuroinflammatory and ischemic injury, yet potentially profibrotic or maladaptive under chronic pathological conditions. This review comprehensively summarizes recent advances in understanding the molecular mechanisms and biological functions of TIMP-1 in CNS diseases, emphasizing its regulatory networks in neuroinflammation, neuroprotection, and neuroregeneration. A deeper understanding of TIMP-1 signaling dynamics will accelerate its translational application as a diagnostic biomarker and therapeutic target for restoring neurovascular unit function in CNS disorders.
Additional Links: PMID-41308758
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308758,
year = {2025},
author = {Zhong, L and Zhao, L and Tang, Q and Zhang, X and Zhou, M and Luo, J and Wang, Y},
title = {Multifunctionality of tissue inhibitor of metalloproteinase-1 in central nervous system diseases.},
journal = {Brain research bulletin},
volume = {},
number = {},
pages = {111647},
doi = {10.1016/j.brainresbull.2025.111647},
pmid = {41308758},
issn = {1873-2747},
abstract = {Central nervous system (CNS) diseases are characterized by high morbidity, long disease courses, and irreversible neurological impairment, often resulting from damage to the neurovascular unit (NVU). Tissue inhibitor of metalloproteinase-1 (TIMP-1), the endogenous inhibitor of matrix metalloproteinase-9 (MMP-9), plays a pivotal role in maintaining extracellular matrix (ECM) homeostasis and regulating NVU integrity. Beyond its canonical MMP-inhibitory function, TIMP-1 exerts a wide spectrum of MMP-independent effects as a multifunctional cytokine that interacts with cell surface receptors such as CD63/β1-integrin and low-density lipoprotein receptor-related protein-1 (LRP-1). Through activation of FAK/PI3K-Akt and MAPK signaling pathways, TIMP-1 modulates astrocyte proliferation, neural stem cell adhesion and migration, endothelial barrier stability, and myelin regeneration. Altered TIMP-1 expression is closely associated with the onset, progression, and prognosis of major CNS disorders, including ischemic stroke, epilepsy, multiple sclerosis, and neurodegenerative diseases such as Alzheimer's and Parkinson's disease. Accumulating evidence highlights its dual, context-dependent roles-protective in acute neuroinflammatory and ischemic injury, yet potentially profibrotic or maladaptive under chronic pathological conditions. This review comprehensively summarizes recent advances in understanding the molecular mechanisms and biological functions of TIMP-1 in CNS diseases, emphasizing its regulatory networks in neuroinflammation, neuroprotection, and neuroregeneration. A deeper understanding of TIMP-1 signaling dynamics will accelerate its translational application as a diagnostic biomarker and therapeutic target for restoring neurovascular unit function in CNS disorders.},
}
RevDate: 2025-11-27
Novel protein acylations in Alzheimer's disease: molecular, mechanisms, biological significance, and diagnostic and therapeutic potentials.
Journal of advanced research pii:S2090-1232(25)00946-4 [Epub ahead of print].
BACKGROUND: Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by complex pathogenesis including amyloid-β (Aβ), hyperphosphorylated tau mediated neurofibrillary tangles (NFTs), energy metabolism disorders, and neuroinflammation, imposing significant burdens on patients' families and society. Increasing evidence implicates epigenetic modifications, particularly novel protein acylations, encompassing endogenous energy metabolites- mediated lysine succinylation (Ksucc), propionylation (Kpr), malonylation (Kmal), crotonylation (Kcr), butyrylation (Kbu), 2-hydroxyisobutyrylation (Khib), β-hydroxybutyrylation (Kbhb), and glutarylation (Kglu), lactylation (Kla), alongside benzoylation (Kbz) mediated by sodium benzoate metabolites and isonicotinylation (Kinic) induced by isoniazid, playing a pivotal role in AD pathogenesis.
AIM OF REVIEW: To enhance the mechanistic understanding of novel acylations, this review systematically summarizes the molecular biological significance of novel protein acylations and their involvement in AD pathogenesis and improvement.
Novel acylations exert profound effects on chromatin architecture, DNA accessibility, and transcriptional regulation. Moreover, they critically coordinate protein properties and functions including modulating protein degradation, protein stability, enzyme activity, protein-protein interaction, and subcellular localization. Dysregulation of specific novel acylations orchestrates key cellular processes such as neuroinflammation, metabolic dysfunction, and programmed cell death, thereby contributing to AD progression. This review systematically delineates the mechanistic foundations of novel acylation modifications and underscores their molecular significance. Furthermore, we comprehensively synthesize current knowledge on the involvement of these acylations in AD, offering novel perspectives for developing targeted preventive and therapeutic strategies.
Additional Links: PMID-41308741
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308741,
year = {2025},
author = {Yang, Y and Yu, Z and Gao, R and Zhang, Y and Zhang, Z and Zhong, X and Jiao, L and Du, K and Wu, Y and Wei, M and Liu, M},
title = {Novel protein acylations in Alzheimer's disease: molecular, mechanisms, biological significance, and diagnostic and therapeutic potentials.},
journal = {Journal of advanced research},
volume = {},
number = {},
pages = {},
doi = {10.1016/j.jare.2025.11.050},
pmid = {41308741},
issn = {2090-1224},
abstract = {BACKGROUND: Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by complex pathogenesis including amyloid-β (Aβ), hyperphosphorylated tau mediated neurofibrillary tangles (NFTs), energy metabolism disorders, and neuroinflammation, imposing significant burdens on patients' families and society. Increasing evidence implicates epigenetic modifications, particularly novel protein acylations, encompassing endogenous energy metabolites- mediated lysine succinylation (Ksucc), propionylation (Kpr), malonylation (Kmal), crotonylation (Kcr), butyrylation (Kbu), 2-hydroxyisobutyrylation (Khib), β-hydroxybutyrylation (Kbhb), and glutarylation (Kglu), lactylation (Kla), alongside benzoylation (Kbz) mediated by sodium benzoate metabolites and isonicotinylation (Kinic) induced by isoniazid, playing a pivotal role in AD pathogenesis.
AIM OF REVIEW: To enhance the mechanistic understanding of novel acylations, this review systematically summarizes the molecular biological significance of novel protein acylations and their involvement in AD pathogenesis and improvement.
Novel acylations exert profound effects on chromatin architecture, DNA accessibility, and transcriptional regulation. Moreover, they critically coordinate protein properties and functions including modulating protein degradation, protein stability, enzyme activity, protein-protein interaction, and subcellular localization. Dysregulation of specific novel acylations orchestrates key cellular processes such as neuroinflammation, metabolic dysfunction, and programmed cell death, thereby contributing to AD progression. This review systematically delineates the mechanistic foundations of novel acylation modifications and underscores their molecular significance. Furthermore, we comprehensively synthesize current knowledge on the involvement of these acylations in AD, offering novel perspectives for developing targeted preventive and therapeutic strategies.},
}
RevDate: 2025-11-27
Default mode network integrity across neuropsychiatric disorders and its relation to social dysfunction: A normative modelling approach.
European neuropsychopharmacology : the journal of the European College of Neuropsychopharmacology, 102:28-38 pii:S0924-977X(25)00781-3 [Epub ahead of print].
Structural and functional default mode network (DMN) alterations are common in neuropsychiatric disorders and may contribute transdiagnostically to social dysfunction. Normative modelling enables assessment of DMN alterations at the individual level. This study investigates whether individual deviations in cortical thickness, surface area, and between-network functional connectivity of the DMN differ between schizophrenia (SZ), major depressive disorder (MDD), Alzheimer's disease (AD), and healthy controls (HC), and whether these deviations transdiagnostically relate to social dysfunction. Social dysfunction was assessed using a composite score from the Social Functioning Scale and De Jong-Gierveld Loneliness scale. Structural MRI data was collected for 329 participants (SZ=86, MDD=44, AD=82, HC=117) and resting-state fMRI data for 317 participants. Individual deviation scores of DMN integrity were computed by adapting existing normative models of cortical thickness (N = 58,836), surface area (N = 43,524), and between-network functional connectivity (N = 21,515). Extreme deviations were quantified using a z-threshold of ±1.96. DMN deviation scores were not transdiagnostically associated with social dysfunction across the sample (ps>0.05). AD patients had more extreme negative deviations in DMN cortical thickness than all other groups (ps<0.0001; z = -4.14 to -6.34) and fewer extreme positive deviations in DMN surface area relative to SZ and HC (ps<0.05; z = 2.10 to 2.71). For between-network functional connectivity of the DMN, AD and SZ patients had more extreme negative deviations than MDD and HC (ps<0.05; z = -2.09 to -3.54). To conclude, normative modelling reveals differences in individual deviations of DMN integrity between neuropsychiatric groups, but these deviations do not transdiagnostically relate to social dysfunction.
Additional Links: PMID-41308510
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308510,
year = {2025},
author = {Braak, S and Oldehinkel, M and Mennes, M and Su, T and Pijnenburg, Y and Arango, C and Ayuso-Mateos, JL and van der Wee, N and Dawson, GR and Marston, HM and Beckmann, CF and Kas, MJH and Penninx, BWJH},
title = {Default mode network integrity across neuropsychiatric disorders and its relation to social dysfunction: A normative modelling approach.},
journal = {European neuropsychopharmacology : the journal of the European College of Neuropsychopharmacology},
volume = {102},
number = {},
pages = {28-38},
doi = {10.1016/j.euroneuro.2025.11.002},
pmid = {41308510},
issn = {1873-7862},
abstract = {Structural and functional default mode network (DMN) alterations are common in neuropsychiatric disorders and may contribute transdiagnostically to social dysfunction. Normative modelling enables assessment of DMN alterations at the individual level. This study investigates whether individual deviations in cortical thickness, surface area, and between-network functional connectivity of the DMN differ between schizophrenia (SZ), major depressive disorder (MDD), Alzheimer's disease (AD), and healthy controls (HC), and whether these deviations transdiagnostically relate to social dysfunction. Social dysfunction was assessed using a composite score from the Social Functioning Scale and De Jong-Gierveld Loneliness scale. Structural MRI data was collected for 329 participants (SZ=86, MDD=44, AD=82, HC=117) and resting-state fMRI data for 317 participants. Individual deviation scores of DMN integrity were computed by adapting existing normative models of cortical thickness (N = 58,836), surface area (N = 43,524), and between-network functional connectivity (N = 21,515). Extreme deviations were quantified using a z-threshold of ±1.96. DMN deviation scores were not transdiagnostically associated with social dysfunction across the sample (ps>0.05). AD patients had more extreme negative deviations in DMN cortical thickness than all other groups (ps<0.0001; z = -4.14 to -6.34) and fewer extreme positive deviations in DMN surface area relative to SZ and HC (ps<0.05; z = 2.10 to 2.71). For between-network functional connectivity of the DMN, AD and SZ patients had more extreme negative deviations than MDD and HC (ps<0.05; z = -2.09 to -3.54). To conclude, normative modelling reveals differences in individual deviations of DMN integrity between neuropsychiatric groups, but these deviations do not transdiagnostically relate to social dysfunction.},
}
RevDate: 2025-11-27
Association Between Omega-3 Supplement Use and Cognitive Function in Korean Older Adults: An 8-Year Longitudinal Cohort Study.
The journal of nutrition, health & aging, 30(1):100739 pii:S1279-7707(25)00264-7 [Epub ahead of print].
OBJECTIVES: South Korea has a relatively high baseline omega-3 fatty acids (O3FA) status due to dietary patterns. However, evidence on additional benefits of O3FA supplementation remains limited. This study examined the 8-year longitudinal effects of O3FA supplement use on cognitive performance among community-dwelling older adults in South Korea.
DESIGN: Prospective longitudinal cohort study.
SETTING AND PARTICIPANTS: Data were from the Korean Longitudinal Study on Cognitive Aging and Dementia (KLOSCAD). A total of 4,949 adults aged ≥60 years (mean age = 69.55 ± 6.56; 44.2% men) were included, and 2,053 completed the 8-year follow-up (2010-2012 to 2018-2020).
MEASUREMENTS: O3FA supplement use was assessed through structured questionnaires on regular intake of omega-3 or fish oil supplements. Cognitive performance was assessed using the Korean version of Consortium to Establish a Registry for Alzheimer's Disease Assessment Packet Neuropsychological Assessment Battery (CERAD-K[N]), and total and memory domain scores were analyzed. Longitudinal changes were examined using repeated measures analysis of covariance and linear mixed-effects models (LMMs; unweighted and weighted), adjusting for potential covariates.
RESULTS: Over the 8 years, CERAD-K[N] (t = -2.686, p = .007) and memory domain (t = -4.026, p < .001) scores significantly improved among O3FA users. In the weighted LMM, significant time × supplement duration interactions were observed (CERAD-K[N]: β = 2.398, 95% CI 1.207-3.589, p < .001; memory: β = 1.050, 95% CI 0.643-1.456, p < .001), indicating greater improvement in long-term users.
CONCLUSION: O3FA supplement use was associated with better maintenance of cognitive function over eight years. These findings suggest a potential role of O3FA supplementation in supporting cognitive health during aging.
Additional Links: PMID-41308405
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308405,
year = {2025},
author = {Lee, WM and Ryu, SI and Lim, MN and Han, JW and Bae, JB and Kim, TH and Kwak, KP and Kim, BJ and Kim, SG and Kim, JL and Moon, SW and Park, JH and Ryu, SH and Lee, DW and Lee, SB and Lee, JJ and Lee, DY and Kim, KW and Jhoo, JH},
title = {Association Between Omega-3 Supplement Use and Cognitive Function in Korean Older Adults: An 8-Year Longitudinal Cohort Study.},
journal = {The journal of nutrition, health & aging},
volume = {30},
number = {1},
pages = {100739},
doi = {10.1016/j.jnha.2025.100739},
pmid = {41308405},
issn = {1760-4788},
abstract = {OBJECTIVES: South Korea has a relatively high baseline omega-3 fatty acids (O3FA) status due to dietary patterns. However, evidence on additional benefits of O3FA supplementation remains limited. This study examined the 8-year longitudinal effects of O3FA supplement use on cognitive performance among community-dwelling older adults in South Korea.
DESIGN: Prospective longitudinal cohort study.
SETTING AND PARTICIPANTS: Data were from the Korean Longitudinal Study on Cognitive Aging and Dementia (KLOSCAD). A total of 4,949 adults aged ≥60 years (mean age = 69.55 ± 6.56; 44.2% men) were included, and 2,053 completed the 8-year follow-up (2010-2012 to 2018-2020).
MEASUREMENTS: O3FA supplement use was assessed through structured questionnaires on regular intake of omega-3 or fish oil supplements. Cognitive performance was assessed using the Korean version of Consortium to Establish a Registry for Alzheimer's Disease Assessment Packet Neuropsychological Assessment Battery (CERAD-K[N]), and total and memory domain scores were analyzed. Longitudinal changes were examined using repeated measures analysis of covariance and linear mixed-effects models (LMMs; unweighted and weighted), adjusting for potential covariates.
RESULTS: Over the 8 years, CERAD-K[N] (t = -2.686, p = .007) and memory domain (t = -4.026, p < .001) scores significantly improved among O3FA users. In the weighted LMM, significant time × supplement duration interactions were observed (CERAD-K[N]: β = 2.398, 95% CI 1.207-3.589, p < .001; memory: β = 1.050, 95% CI 0.643-1.456, p < .001), indicating greater improvement in long-term users.
CONCLUSION: O3FA supplement use was associated with better maintenance of cognitive function over eight years. These findings suggest a potential role of O3FA supplementation in supporting cognitive health during aging.},
}
RevDate: 2025-11-27
Identification and validation of PANoptosis-related genes in Alzheimer's disease.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
BackgroundAlzheimer's disease (AD) is the main cause of dementia in older adults. Recently, increasing evidence shows that PANoptosis plays an important role in AD.ObjectiveThis study investigated potential roles of PANoptosis by bioinformatics and machine learning in AD.MethodsAD-related microarray sets were downloaded from the GEO database and PANoptosis-related genes were extracted from the GeneCards database. By WGCNA and constructing machine learning models, hub genes were identified and verified. A ceRNA network was established using cytoscape. The ssGSEA was used to estimate immune cell infiltration and its correlation with hub genes. The R package was performed for consensus clustering (CC) analysis.Results240 differentially expressed genes in the training set were identified. By selecting optimal models, we finally identified five PANoptosis-related hub genes in AD: ADCYAP1, BCL6, CXCR4, SPP1, and PGF, which were verified in the validation set (excluding SPP1 unverified) and the Aβ25-35-induced AD cell model. Subsequently, a risk prediction model with good performance for AD and a ceRNA network was established. Then, it was found that 14 types of immune cells with increased expression and 5 types with decreased expression in AD, significantly related to hub genes. Finally, two AD subtypes were proposed based on CC analysis: high immune infiltrative (more immune cell expression associated with inflammation and programmed cell death pathways) and low immune infiltrative subtype.ConclusionsOur results suggest that five PANoptosis-related genes are significantly associated with the pathologic progression of AD; we proposed two AD immune infiltrative subtypes.
Additional Links: PMID-41308018
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308018,
year = {2025},
author = {Zhou, W and Lin, H and Huang, X and Zhou, X},
title = {Identification and validation of PANoptosis-related genes in Alzheimer's disease.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251396927},
doi = {10.1177/13872877251396927},
pmid = {41308018},
issn = {1875-8908},
abstract = {BackgroundAlzheimer's disease (AD) is the main cause of dementia in older adults. Recently, increasing evidence shows that PANoptosis plays an important role in AD.ObjectiveThis study investigated potential roles of PANoptosis by bioinformatics and machine learning in AD.MethodsAD-related microarray sets were downloaded from the GEO database and PANoptosis-related genes were extracted from the GeneCards database. By WGCNA and constructing machine learning models, hub genes were identified and verified. A ceRNA network was established using cytoscape. The ssGSEA was used to estimate immune cell infiltration and its correlation with hub genes. The R package was performed for consensus clustering (CC) analysis.Results240 differentially expressed genes in the training set were identified. By selecting optimal models, we finally identified five PANoptosis-related hub genes in AD: ADCYAP1, BCL6, CXCR4, SPP1, and PGF, which were verified in the validation set (excluding SPP1 unverified) and the Aβ25-35-induced AD cell model. Subsequently, a risk prediction model with good performance for AD and a ceRNA network was established. Then, it was found that 14 types of immune cells with increased expression and 5 types with decreased expression in AD, significantly related to hub genes. Finally, two AD subtypes were proposed based on CC analysis: high immune infiltrative (more immune cell expression associated with inflammation and programmed cell death pathways) and low immune infiltrative subtype.ConclusionsOur results suggest that five PANoptosis-related genes are significantly associated with the pathologic progression of AD; we proposed two AD immune infiltrative subtypes.},
}
RevDate: 2025-11-27
Identification of common human TMEM173 genotypes associated with Alzheimer's disease.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
BackgroundRecent animal studies have revealed STING (Stimulator of interferon genes) as a potential key player in Alzheimer's disease (AD). The actual impact of human STING on AD, however, is unknown. Mouse STING studies were done in WT/WT. However, TMEM173, the human gene encodes STING, has 5 common, distinct, sometimes opposite functional alleles that result in 25 TMEM173 genotypes. Only ∼50% of whites, 36% of African Americans (AA), 22% of East Asians are WT/WT. Past STING cancer immunotherapy clinic trials, which did not consider human TMEM173 heterogeneity, all failed.Objective(1) Discover new protective and risk AD genetic factors across populations or AA-specific. (2) Establish the physiological significance of common human TMEM173 genotypes and human diseases.MethodsWe conduct a large-scale (∼15,000 individuals) case-control analysis between TMEM173 genotypes and AD using data from The National Institute on Aging Genetics of Alzheimer's Disease Data Storage Site. The data include late-onset AD (LOAD) non-Hispanic White (NHW), early-onset AD (EOAD) NHW, and AA.ResultsA common H232/HAQ TMEM173 genotype is associated with AD protection across the populations. An AA-specific TMEM173 genotype H232/Q293 increases the risk for AA males (OR = 17.7148), especially in the APOE ε3/ε3 population.ConclusionsThe findings discovered the first AA-specific high AD risk factor and established an association between human TMEM173 and AD, paving the way for STING-targeting effective AD healthcare.
Additional Links: PMID-41308015
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308015,
year = {2025},
author = {Usher, MR and Aybar-Torres, AA and Jin, L},
title = {Identification of common human TMEM173 genotypes associated with Alzheimer's disease.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251396973},
doi = {10.1177/13872877251396973},
pmid = {41308015},
issn = {1875-8908},
abstract = {BackgroundRecent animal studies have revealed STING (Stimulator of interferon genes) as a potential key player in Alzheimer's disease (AD). The actual impact of human STING on AD, however, is unknown. Mouse STING studies were done in WT/WT. However, TMEM173, the human gene encodes STING, has 5 common, distinct, sometimes opposite functional alleles that result in 25 TMEM173 genotypes. Only ∼50% of whites, 36% of African Americans (AA), 22% of East Asians are WT/WT. Past STING cancer immunotherapy clinic trials, which did not consider human TMEM173 heterogeneity, all failed.Objective(1) Discover new protective and risk AD genetic factors across populations or AA-specific. (2) Establish the physiological significance of common human TMEM173 genotypes and human diseases.MethodsWe conduct a large-scale (∼15,000 individuals) case-control analysis between TMEM173 genotypes and AD using data from The National Institute on Aging Genetics of Alzheimer's Disease Data Storage Site. The data include late-onset AD (LOAD) non-Hispanic White (NHW), early-onset AD (EOAD) NHW, and AA.ResultsA common H232/HAQ TMEM173 genotype is associated with AD protection across the populations. An AA-specific TMEM173 genotype H232/Q293 increases the risk for AA males (OR = 17.7148), especially in the APOE ε3/ε3 population.ConclusionsThe findings discovered the first AA-specific high AD risk factor and established an association between human TMEM173 and AD, paving the way for STING-targeting effective AD healthcare.},
}
RevDate: 2025-11-27
Hub genes and diagnostic model associated with mitochondrial function in Alzheimer's disease.
Animal models and experimental medicine [Epub ahead of print].
BACKGROUND: Alzheimer's disease (AD) represents the most prevalent neurodegenerative disorder, with mitochondrial dysfunction being observed in both AD patients and mouse models. Nonetheless, further investigation is required to elucidate the pathogenic genes associated with AD and to develop early diagnostic methodologies centered on mitochondrial function.
METHODS: In this study, the dataset GSE132903 was retrieved from the GEO database, encompassing both non-demented (ND) control and AD samples. Through the combination of differential expression gene analysis, weighted gene co-expression network analysis, and intersection with mitochondrial database gene sets, four hub genes associated with AD were identified. These four hub genes were subsequently validated in APP/PS1 and 5xFAD mouse models using molecular biology techniques.
RESULTS: The hub genes identified through bioinformatics analysis include SYNJ2BP, VDAC1, NUBPL, and COX19. Within the GSE132903 dataset, the expression levels of SYNJ2BP, NUBPL, and COX19 were significantly elevated in the AD group compared to the non-demented (ND) group, whereas VDAC1 expression was reduced in the AD group relative to the ND group. Furthermore, in the hippocampus of APP/PS1 and 5xFAD mouse models, the expression patterns of SYNJ2BP and NUBPL were consistent with the bioinformatics analysis results.
CONCLUSION: Hub genes identified here through bioinformatics and molecular biology may help early diagnosis of AD patients and may also help build new AD models to explore its pathogenesis.
Additional Links: PMID-41308013
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41308013,
year = {2025},
author = {Zhu, X and Zhang, L and Qin, C},
title = {Hub genes and diagnostic model associated with mitochondrial function in Alzheimer's disease.},
journal = {Animal models and experimental medicine},
volume = {},
number = {},
pages = {},
doi = {10.1002/ame2.70104},
pmid = {41308013},
issn = {2576-2095},
support = {2023-PT180-01//Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences/ ; 2023-PT330-01//Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences/ ; 2021-I2M-1-034//Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences/ ; 82161138027//National Natural Science Foundation of China/ ; },
abstract = {BACKGROUND: Alzheimer's disease (AD) represents the most prevalent neurodegenerative disorder, with mitochondrial dysfunction being observed in both AD patients and mouse models. Nonetheless, further investigation is required to elucidate the pathogenic genes associated with AD and to develop early diagnostic methodologies centered on mitochondrial function.
METHODS: In this study, the dataset GSE132903 was retrieved from the GEO database, encompassing both non-demented (ND) control and AD samples. Through the combination of differential expression gene analysis, weighted gene co-expression network analysis, and intersection with mitochondrial database gene sets, four hub genes associated with AD were identified. These four hub genes were subsequently validated in APP/PS1 and 5xFAD mouse models using molecular biology techniques.
RESULTS: The hub genes identified through bioinformatics analysis include SYNJ2BP, VDAC1, NUBPL, and COX19. Within the GSE132903 dataset, the expression levels of SYNJ2BP, NUBPL, and COX19 were significantly elevated in the AD group compared to the non-demented (ND) group, whereas VDAC1 expression was reduced in the AD group relative to the ND group. Furthermore, in the hippocampus of APP/PS1 and 5xFAD mouse models, the expression patterns of SYNJ2BP and NUBPL were consistent with the bioinformatics analysis results.
CONCLUSION: Hub genes identified here through bioinformatics and molecular biology may help early diagnosis of AD patients and may also help build new AD models to explore its pathogenesis.},
}
RevDate: 2025-11-27
Spatial and single-cell transcriptomics reveal the reorganization of cerebellar microglia with aging.
Cell reports, 44(12):116624 pii:S2211-1247(25)01396-8 [Epub ahead of print].
The cerebellum, essential for motor coordination and increasingly recognized for its role in cognition, is typically considered more resilient to aging and largely spared from hallmark Alzheimer's disease (AD) pathology. However, transcriptomic analyses across fifteen mouse brain regions revealed that the cerebellum undergoes some of the earliest and most pronounced age-related changes. To investigate cerebellar aging, we applied single-nucleus RNA sequencing (RNA-seq), microglial bulk RNA-seq, and multiplexed error-robust fluorescence in situ hybridization (MERFISH)-based spatial transcriptomics. Microglia showed the most prominent changes, including elevated expression of a neuroprotective signature and reduced expression of a lipid-droplet-accumulating signature compared to hippocampal microglia. Spatial analyses further revealed that aged cerebellar microglia were positioned in close proximity to granule cells. Utilizing this relationship, we identified a proximity-dependent transcriptional state defined by the neuron-associated microglial signature. This signature reveals a region-specific microglial adaptation, highlighting cerebellar reorganization with age and potential resilience to AD.
Additional Links: PMID-41307999
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307999,
year = {2025},
author = {Tsai, AP and Henze, DE and Ramirez Lopez, E and Haberberger, J and Dong, C and Lu, N and Atkins, M and Costa, EK and Farinas, A and Oh, HS and Moran-Losada, P and Le Guen, Y and Isakova, A and Quake, SR and Wyss-Coray, T},
title = {Spatial and single-cell transcriptomics reveal the reorganization of cerebellar microglia with aging.},
journal = {Cell reports},
volume = {44},
number = {12},
pages = {116624},
doi = {10.1016/j.celrep.2025.116624},
pmid = {41307999},
issn = {2211-1247},
abstract = {The cerebellum, essential for motor coordination and increasingly recognized for its role in cognition, is typically considered more resilient to aging and largely spared from hallmark Alzheimer's disease (AD) pathology. However, transcriptomic analyses across fifteen mouse brain regions revealed that the cerebellum undergoes some of the earliest and most pronounced age-related changes. To investigate cerebellar aging, we applied single-nucleus RNA sequencing (RNA-seq), microglial bulk RNA-seq, and multiplexed error-robust fluorescence in situ hybridization (MERFISH)-based spatial transcriptomics. Microglia showed the most prominent changes, including elevated expression of a neuroprotective signature and reduced expression of a lipid-droplet-accumulating signature compared to hippocampal microglia. Spatial analyses further revealed that aged cerebellar microglia were positioned in close proximity to granule cells. Utilizing this relationship, we identified a proximity-dependent transcriptional state defined by the neuron-associated microglial signature. This signature reveals a region-specific microglial adaptation, highlighting cerebellar reorganization with age and potential resilience to AD.},
}
RevDate: 2025-11-27
Early axonal degeneration linked to clinical decline in Alzheimer's disease progression revealed with diffusion MRI.
The Journal of clinical investigation pii:196638 [Epub ahead of print].
BACKGROUND: Axonal degeneration is believed to be an early hallmark of Alzheimer's disease (AD). This study investigated the temporal trajectory of axonal loss and its association with cognitive and functional decline using diffusion MRI-derived Axonal Density Index (dMRI-ADI).
METHODS: Longitudinal dMRI, CSF and PET data from the ADNI were analyzed, including 117 cognitively normal (CN) and 88 impaired (CI) subjects, consisting of 74 mild cognitive impairment (MCI) and 14 AD individuals. Linear mixed-effects models examined group differences as well as associations between baseline and longitudinal changes in ADI, CSF or PET biomarkers and clinical outcomes. Results derived from larger CSF (n=527) and PET (tau-PET: n=870; amyloid-PET: n=1581) data were also presented.
RESULTS: Compared to CN, the CI group exhibited significantly lower baseline ADI values and steeper longitudinal decline (p<10-⁶). Lower baseline ADI predicted faster cognitive and functional decline in the CI group (MMSE: p=0.03; CDR-SB: p<10-⁴), and longitudinal decreases in ADI were associated with worsening clinical outcomes (MMSE: p=0.001; CDR-SB: p<10-¹²). Compared to CSF and PET biomarkers, ADI demonstrated superior sensitivity in tracking disease progression and matched these biomarkers in predicting future cognitive and functional decline. Furthermore, decreases in ADI were significantly associated with declines in clinical outcomes; an association observed only with amyloid-PET, but not CSF biomarkers.
CONCLUSION: Axonal degeneration is an early and clinically meaningful feature of AD. ADI is a promising noninvasive biomarker for early detection, prognosis, and disease monitoring.
CLINICALTRIALS: gov NCT00106899.
FUNDING: This work was supported by the National Institute on Aging IRP.
Additional Links: PMID-41307953
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307953,
year = {2025},
author = {Gong, Z and Laporte, JP and Guo, AY and Bilgel, M and Bae, J and Fox, NY and de Rouen, A and Zhang, N and Taranath, A and de Cabo, R and Egan, JM and Ferrucci, L and Bouhrara, M},
title = {Early axonal degeneration linked to clinical decline in Alzheimer's disease progression revealed with diffusion MRI.},
journal = {The Journal of clinical investigation},
volume = {},
number = {},
pages = {},
doi = {10.1172/JCI196638},
pmid = {41307953},
issn = {1558-8238},
abstract = {BACKGROUND: Axonal degeneration is believed to be an early hallmark of Alzheimer's disease (AD). This study investigated the temporal trajectory of axonal loss and its association with cognitive and functional decline using diffusion MRI-derived Axonal Density Index (dMRI-ADI).
METHODS: Longitudinal dMRI, CSF and PET data from the ADNI were analyzed, including 117 cognitively normal (CN) and 88 impaired (CI) subjects, consisting of 74 mild cognitive impairment (MCI) and 14 AD individuals. Linear mixed-effects models examined group differences as well as associations between baseline and longitudinal changes in ADI, CSF or PET biomarkers and clinical outcomes. Results derived from larger CSF (n=527) and PET (tau-PET: n=870; amyloid-PET: n=1581) data were also presented.
RESULTS: Compared to CN, the CI group exhibited significantly lower baseline ADI values and steeper longitudinal decline (p<10-⁶). Lower baseline ADI predicted faster cognitive and functional decline in the CI group (MMSE: p=0.03; CDR-SB: p<10-⁴), and longitudinal decreases in ADI were associated with worsening clinical outcomes (MMSE: p=0.001; CDR-SB: p<10-¹²). Compared to CSF and PET biomarkers, ADI demonstrated superior sensitivity in tracking disease progression and matched these biomarkers in predicting future cognitive and functional decline. Furthermore, decreases in ADI were significantly associated with declines in clinical outcomes; an association observed only with amyloid-PET, but not CSF biomarkers.
CONCLUSION: Axonal degeneration is an early and clinically meaningful feature of AD. ADI is a promising noninvasive biomarker for early detection, prognosis, and disease monitoring.
CLINICALTRIALS: gov NCT00106899.
FUNDING: This work was supported by the National Institute on Aging IRP.},
}
RevDate: 2025-11-27
Care Partners' Perceptions of Amyloid-Targeting Therapy and Treat‑to‑Clearance for Alzheimer's Disease in Japan: A Qualitative Study.
Neurology and therapy [Epub ahead of print].
INTRODUCTION: Donanemab has been developed as an amyloid-targeting therapy (ATT) for mild cognitive impairment (MCI) and mild dementia due to Alzheimer's disease (AD). In registration trials involving donanemab, a treat‑to‑clearance approach was used, in which patients discontinued ATT when amyloid plaque levels decreased below a predefined threshold, which differs from previously available symptomatic treatments for AD. Our study explored care partners' perceptions regarding ATT and treat‑to‑clearance.
METHODS: This was a cross-sectional, qualitative interview study. Care partners of individuals with MCI or mild dementia due to AD participated in online semi-structured interviews about their perceptions regarding the impact of MCI or mild dementia diagnoses due to AD, the burden of supporting, and use/cessation of ATT. The qualitative data from the interviews were analyzed using a thematic approach.
RESULTS: The participants were 22 care partners (5 male/17 female), and their median age was 59 (range 35-81) years. The most common relationships between care partners and the individuals with AD were child (50.0%) and spouse/partner (45.5%); 68.2% of the care partners lived with the individuals with AD. Thematic analysis identified three major classifications (Thoughts regarding therapy; Treat‑to‑clearance; and Burdens of support), along with 15 themes and five sub-themes. Care partners expressed experiencing mental burden and time constraints, while treat‑to‑clearance could save care partners' time by reducing hospital waiting time and alleviating financial burden. Confirming the clearance of amyloid β plaques provided care partners with a sense of relief, while they remained concerned about the potential progression of AD symptoms and sought follow-up care after stopping treatment.
CONCLUSIONS: These results suggest that providing clear explanations and facilitating shared decision-making when introducing ATT, as well as introducing follow-up care and long-term evidence after stopping treatment, are needed.
Additional Links: PMID-41307609
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307609,
year = {2025},
author = {Katayama, S and Tsujimoto, M and Suzuki, K and Ueda, K and Shimoura, K and Suo, S and Hatakeyama, N},
title = {Care Partners' Perceptions of Amyloid-Targeting Therapy and Treat‑to‑Clearance for Alzheimer's Disease in Japan: A Qualitative Study.},
journal = {Neurology and therapy},
volume = {},
number = {},
pages = {},
pmid = {41307609},
issn = {2193-8253},
abstract = {INTRODUCTION: Donanemab has been developed as an amyloid-targeting therapy (ATT) for mild cognitive impairment (MCI) and mild dementia due to Alzheimer's disease (AD). In registration trials involving donanemab, a treat‑to‑clearance approach was used, in which patients discontinued ATT when amyloid plaque levels decreased below a predefined threshold, which differs from previously available symptomatic treatments for AD. Our study explored care partners' perceptions regarding ATT and treat‑to‑clearance.
METHODS: This was a cross-sectional, qualitative interview study. Care partners of individuals with MCI or mild dementia due to AD participated in online semi-structured interviews about their perceptions regarding the impact of MCI or mild dementia diagnoses due to AD, the burden of supporting, and use/cessation of ATT. The qualitative data from the interviews were analyzed using a thematic approach.
RESULTS: The participants were 22 care partners (5 male/17 female), and their median age was 59 (range 35-81) years. The most common relationships between care partners and the individuals with AD were child (50.0%) and spouse/partner (45.5%); 68.2% of the care partners lived with the individuals with AD. Thematic analysis identified three major classifications (Thoughts regarding therapy; Treat‑to‑clearance; and Burdens of support), along with 15 themes and five sub-themes. Care partners expressed experiencing mental burden and time constraints, while treat‑to‑clearance could save care partners' time by reducing hospital waiting time and alleviating financial burden. Confirming the clearance of amyloid β plaques provided care partners with a sense of relief, while they remained concerned about the potential progression of AD symptoms and sought follow-up care after stopping treatment.
CONCLUSIONS: These results suggest that providing clear explanations and facilitating shared decision-making when introducing ATT, as well as introducing follow-up care and long-term evidence after stopping treatment, are needed.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
The (un-)Social Brain in Isolation.
Physiological research, 74(5):711-727.
The Social Brain is a distributed network of neuroanatomical regions and neurochemical systems that underpins the human capacity for social cognition, empathy, and interpersonal behavior. Social isolation (SI), defined as the objective reduction in social interaction, poses a significant threat to the integrity of this system. In this review, we synthesize evidence from human and animal studies to elucidate the biological, cognitive, and behavioral consequences of SI on the social brain. We describe how SI acts as a chronic stressor, disrupting structural connectivity, and altering neurotransmitter systems critical for social cognition. These disruptions manifest in altered social behavior, mentalization processes, and emotional reactivity, significantly contributing to increased vulnerability to psychiatric and neurodegenerative disorders, including depression, schizophrenia, substance use disorders, and Alzheimer's disease. Converging findings from studies of evolutionarily conserved mechanisms in rodent and primate models demonstrate that SI compromises neurodevelopment, attenuates neuroplasticity, and triggers maladaptive stress responses, highlighting that social deprivation has profound neurobiological and behavioral consequences that greatly overlap with the pathophysiological changes seen in neuropsychiatric disorders. Furthermore, we explore the role of indirect stressors resulting from SI such as touch deprivation and digital-era social disconnection as contemporary amplifiers of SI's neurobiological impact. In light of public health challenges such as the COVID-19 pandemic, we propose that SI should be recognized not only as a psychosocial condition but as a modifiable risk factor with transdiagnostic significance across psychiatry, neurology, and preventive medicine. Addressing SI through targeted interventions and policy measures is essential for promoting mental resilience and well-being. Key words Chronic Stress " Loneliness " Social Cognition " Socialization " Social Stress.
Additional Links: PMID-41307506
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307506,
year = {2025},
author = {Fishman, A and Grinin, P and Riljak, V},
title = {The (un-)Social Brain in Isolation.},
journal = {Physiological research},
volume = {74},
number = {5},
pages = {711-727},
pmid = {41307506},
issn = {1802-9973},
mesh = {Humans ; *Social Isolation/psychology ; *Brain/physiology/physiopathology ; Animals ; *COVID-19/psychology ; *Social Behavior ; *Stress, Psychological/psychology ; },
abstract = {The Social Brain is a distributed network of neuroanatomical regions and neurochemical systems that underpins the human capacity for social cognition, empathy, and interpersonal behavior. Social isolation (SI), defined as the objective reduction in social interaction, poses a significant threat to the integrity of this system. In this review, we synthesize evidence from human and animal studies to elucidate the biological, cognitive, and behavioral consequences of SI on the social brain. We describe how SI acts as a chronic stressor, disrupting structural connectivity, and altering neurotransmitter systems critical for social cognition. These disruptions manifest in altered social behavior, mentalization processes, and emotional reactivity, significantly contributing to increased vulnerability to psychiatric and neurodegenerative disorders, including depression, schizophrenia, substance use disorders, and Alzheimer's disease. Converging findings from studies of evolutionarily conserved mechanisms in rodent and primate models demonstrate that SI compromises neurodevelopment, attenuates neuroplasticity, and triggers maladaptive stress responses, highlighting that social deprivation has profound neurobiological and behavioral consequences that greatly overlap with the pathophysiological changes seen in neuropsychiatric disorders. Furthermore, we explore the role of indirect stressors resulting from SI such as touch deprivation and digital-era social disconnection as contemporary amplifiers of SI's neurobiological impact. In light of public health challenges such as the COVID-19 pandemic, we propose that SI should be recognized not only as a psychosocial condition but as a modifiable risk factor with transdiagnostic significance across psychiatry, neurology, and preventive medicine. Addressing SI through targeted interventions and policy measures is essential for promoting mental resilience and well-being. Key words Chronic Stress " Loneliness " Social Cognition " Socialization " Social Stress.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Social Isolation/psychology
*Brain/physiology/physiopathology
Animals
*COVID-19/psychology
*Social Behavior
*Stress, Psychological/psychology
RevDate: 2025-11-27
CmpDate: 2025-11-27
Dipeptidyl Peptidase 4 Mediated Caspase-8 Affects Cognitive Impairment in Mice With Alzheimer's Disease.
FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 39(23):e71264.
To investigate the effect of dipeptidyl peptidase 4 (DPP4) on cognitive impairment in Alzheimer's disease (AD), the present study used seven-week-old male C57BL/6J and DPP4 knockout mice. The AD model was induced by microinjection of Aβ25-35 into the lateral ventricle. Morris water maze test showed that DPP4 knockout significantly improved the spatial learning and memory abilities of AD mice. Western blot results showed that DPP4 knockout increased the expression levels of BDNF, CREB and Bcl-2 in the hippocampus of AD mice while the expression levels of Caspase-8, pyroptosis-related proteins NLRP3, Caspase-1, GSDMD, IL-118, IL-1β, and apoptosis-related proteins Caspase-3 and Bax were decreased. Similar results were observed after HT22 neurons were treated with Aβ25-35 and the DPP4 inhibitor sitagliptin (Sit). Moreover, the treatment with a Caspase-8 inhibitor (Z-LETD-FMK) showed that the inhibition of Caspase-8 inhibited the expression of NLRP3 and Caspase-1 in the AD model cells, but had no further inhibitory effect under the treatment of Sit. Our results suggest that DPP4 knockout may ameliorate learning and memory dysfunction in AD model mice by regulating pyroptosis and apoptosis pathways through Caspase-8.
Additional Links: PMID-41307478
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307478,
year = {2025},
author = {Wang, X and Chen, L and Qiu, J and Wang, K and Xi, P and Cheng, X and He, Z and Jiang, H},
title = {Dipeptidyl Peptidase 4 Mediated Caspase-8 Affects Cognitive Impairment in Mice With Alzheimer's Disease.},
journal = {FASEB journal : official publication of the Federation of American Societies for Experimental Biology},
volume = {39},
number = {23},
pages = {e71264},
doi = {10.1096/fj.202501322RR},
pmid = {41307478},
issn = {1530-6860},
support = {82204837//MOST | NSFC | National Natural Science Foundation of China-Zhejiang Joint Fund for the Integration of Industrialization and Informatization (NSFC-Zhejiang Joint Fund)/ ; 81760207//MOST | NSFC | National Natural Science Foundation of China-Zhejiang Joint Fund for the Integration of Industrialization and Informatization (NSFC-Zhejiang Joint Fund)/ ; LQ23H290004//Zhejiang Provincial Natural Science Foundation of China/ ; 2024AY10021//Jiaxing Science and Technology Plan Project/ ; },
mesh = {Animals ; *Alzheimer Disease/metabolism ; Male ; Mice ; *Dipeptidyl Peptidase 4/metabolism/genetics ; Mice, Inbred C57BL ; *Caspase 8/metabolism/genetics ; *Cognitive Dysfunction/metabolism ; Mice, Knockout ; Amyloid beta-Peptides/toxicity ; Disease Models, Animal ; Pyroptosis ; Dipeptidyl-Peptidase IV Inhibitors/pharmacology ; Maze Learning ; Hippocampus/metabolism ; Neurons/metabolism ; },
abstract = {To investigate the effect of dipeptidyl peptidase 4 (DPP4) on cognitive impairment in Alzheimer's disease (AD), the present study used seven-week-old male C57BL/6J and DPP4 knockout mice. The AD model was induced by microinjection of Aβ25-35 into the lateral ventricle. Morris water maze test showed that DPP4 knockout significantly improved the spatial learning and memory abilities of AD mice. Western blot results showed that DPP4 knockout increased the expression levels of BDNF, CREB and Bcl-2 in the hippocampus of AD mice while the expression levels of Caspase-8, pyroptosis-related proteins NLRP3, Caspase-1, GSDMD, IL-118, IL-1β, and apoptosis-related proteins Caspase-3 and Bax were decreased. Similar results were observed after HT22 neurons were treated with Aβ25-35 and the DPP4 inhibitor sitagliptin (Sit). Moreover, the treatment with a Caspase-8 inhibitor (Z-LETD-FMK) showed that the inhibition of Caspase-8 inhibited the expression of NLRP3 and Caspase-1 in the AD model cells, but had no further inhibitory effect under the treatment of Sit. Our results suggest that DPP4 knockout may ameliorate learning and memory dysfunction in AD model mice by regulating pyroptosis and apoptosis pathways through Caspase-8.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Alzheimer Disease/metabolism
Male
Mice
*Dipeptidyl Peptidase 4/metabolism/genetics
Mice, Inbred C57BL
*Caspase 8/metabolism/genetics
*Cognitive Dysfunction/metabolism
Mice, Knockout
Amyloid beta-Peptides/toxicity
Disease Models, Animal
Pyroptosis
Dipeptidyl-Peptidase IV Inhibitors/pharmacology
Maze Learning
Hippocampus/metabolism
Neurons/metabolism
RevDate: 2025-11-27
Pharmaceutical applications of cyclopropyl containing scaffolds: a review on recent updates.
Future medicinal chemistry [Epub ahead of print].
Carbocycles have been widely employed in the development of pharmaceutically active scaffolds. Cyclopropane has attracted significant attention from researchers due to its unique chemical properties among carbocycles. Subsequently, this review will focus on cyclopropane-containing pharmaceutical drug products that have been approved by the FDA (Food and Drug Administration) and are used to treat a wide variety of medical conditions. In addition to the synthesis of the cyclopropyl moiety through various chemical reactions, such as the Corey-Chaykovsky reaction and the Simmons-Smith reaction. Several cyclopropane-containing pharmaceutical drugs have been reported to exert significant anti-coagulant effects. Additionally, they also exhibit inhibitory activity against MET, a receptor tyrosine kinase, as well as vascular endothelial growth factor receptor 2 (VEGFR-2). Moreover, they showed cytotoxicity by inhibiting epidermal growth factor receptor (EGFR[L858R/T790M]). In addition to antidiabetic, anti-Alzheimer, antimalarial, antimicrobial, anti-convulsant and anti-depressant activities. Herein, we present the pharmaceutical applications of cyclopropane-containing derivatives, shedding light on the structure-activity relationship (SAR), along with some commonly reported methods for their synthesis.
Additional Links: PMID-41307432
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307432,
year = {2025},
author = {Nemr, MTM and Elshaier, YAAM and Ewieda, SY and Abdelaziz, MA},
title = {Pharmaceutical applications of cyclopropyl containing scaffolds: a review on recent updates.},
journal = {Future medicinal chemistry},
volume = {},
number = {},
pages = {1-10},
doi = {10.1080/17568919.2025.2594969},
pmid = {41307432},
issn = {1756-8927},
abstract = {Carbocycles have been widely employed in the development of pharmaceutically active scaffolds. Cyclopropane has attracted significant attention from researchers due to its unique chemical properties among carbocycles. Subsequently, this review will focus on cyclopropane-containing pharmaceutical drug products that have been approved by the FDA (Food and Drug Administration) and are used to treat a wide variety of medical conditions. In addition to the synthesis of the cyclopropyl moiety through various chemical reactions, such as the Corey-Chaykovsky reaction and the Simmons-Smith reaction. Several cyclopropane-containing pharmaceutical drugs have been reported to exert significant anti-coagulant effects. Additionally, they also exhibit inhibitory activity against MET, a receptor tyrosine kinase, as well as vascular endothelial growth factor receptor 2 (VEGFR-2). Moreover, they showed cytotoxicity by inhibiting epidermal growth factor receptor (EGFR[L858R/T790M]). In addition to antidiabetic, anti-Alzheimer, antimalarial, antimicrobial, anti-convulsant and anti-depressant activities. Herein, we present the pharmaceutical applications of cyclopropane-containing derivatives, shedding light on the structure-activity relationship (SAR), along with some commonly reported methods for their synthesis.},
}
RevDate: 2025-11-27
Molecular pathways underlying amyloid precursor protein-mediated regulation of adult-born neurons.
Neural regeneration research pii:01300535-990000000-01057 [Epub ahead of print].
Cleavage of amyloid precursor protein (APP) produces toxic amyloid-beta peptides, which play a critical role in the pathogenesis of Alzheimer's disease. Neuronal loss is a key feature of Alzheimer's disease. Despite the importance of APP in the proliferation of neural progenitors and the survival of adult-born granule cells in the dentate gyrus, little is known about the effect of APP deficiency on neuronal electrophysiological activities and the survival of newly born neurons. Utilizing whole-cell patch-clamp recording in combination with retroviral labeling and immunofluorescent staining in Alzheimer's disease model mice with App knockout (App-/-), we show that APP deficiency increased the number of adult-born granule cells at 4 weeks post-injection, but did not affect their intrinsic excitability or miniature current activity. In contrast, at 10 weeks post-injection, adult-born granule cells showed increased abundance and intrinsic excitability that were associated with abnormal dendritic morphology, increased miniature excitatory- and inhibitory-synaptic transmission, and decreased potassium-chloride-cotransporter 2 expression. Compared with adult-born granule cells at 10 week post-injection, mature granule cells exhibited decreased intrinsic excitability and potassium-chloride-cotransporter 2 expression alongside increased apoptosis in App-/- mice. Additionally, although App-/-mice showed abnormal freezing behavior and elevated mature granule cell activation during contextual fear conditioning, adult-born granule cells were not recruited in either App-/- or wild-type control mice. Taken together, these findings suggest that APP is required for adult-born granule cell maturation and that APP deficiency induces excitotoxicity in adult-born granule cells at 10 weeks post-injection, promoting subsequent apoptosis of mature granule cells.
Additional Links: PMID-41307255
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307255,
year = {2025},
author = {Hu, H and Li, H and Chen, Y and Zhou, J and Chen, J and Tang, Q and Chen, X and Jiang, J and Sun, M and Jia, D and Xie, W and Long, C and Yang, L},
title = {Molecular pathways underlying amyloid precursor protein-mediated regulation of adult-born neurons.},
journal = {Neural regeneration research},
volume = {},
number = {},
pages = {},
doi = {10.4103/NRR.NRR-D-25-00175},
pmid = {41307255},
issn = {1673-5374},
abstract = {Cleavage of amyloid precursor protein (APP) produces toxic amyloid-beta peptides, which play a critical role in the pathogenesis of Alzheimer's disease. Neuronal loss is a key feature of Alzheimer's disease. Despite the importance of APP in the proliferation of neural progenitors and the survival of adult-born granule cells in the dentate gyrus, little is known about the effect of APP deficiency on neuronal electrophysiological activities and the survival of newly born neurons. Utilizing whole-cell patch-clamp recording in combination with retroviral labeling and immunofluorescent staining in Alzheimer's disease model mice with App knockout (App-/-), we show that APP deficiency increased the number of adult-born granule cells at 4 weeks post-injection, but did not affect their intrinsic excitability or miniature current activity. In contrast, at 10 weeks post-injection, adult-born granule cells showed increased abundance and intrinsic excitability that were associated with abnormal dendritic morphology, increased miniature excitatory- and inhibitory-synaptic transmission, and decreased potassium-chloride-cotransporter 2 expression. Compared with adult-born granule cells at 10 week post-injection, mature granule cells exhibited decreased intrinsic excitability and potassium-chloride-cotransporter 2 expression alongside increased apoptosis in App-/- mice. Additionally, although App-/-mice showed abnormal freezing behavior and elevated mature granule cell activation during contextual fear conditioning, adult-born granule cells were not recruited in either App-/- or wild-type control mice. Taken together, these findings suggest that APP is required for adult-born granule cell maturation and that APP deficiency induces excitotoxicity in adult-born granule cells at 10 weeks post-injection, promoting subsequent apoptosis of mature granule cells.},
}
RevDate: 2025-11-27
Hippocampal neural stem cell-derived extracellular vesicles modulate microglia to promote resilience against tau oligomers.
Neural regeneration research pii:01300535-990000000-01072 [Epub ahead of print].
Neural stem cells and adult hippocampal neurogenesis modulate synaptic plasticity and cognitive function. Neural stem cells secrete extracellular vesicles - microvesicles carrying biomolecular cargos - that modulate the function of other cells and contribute to homeostasis and plasticity in the central nervous system. Alzheimer's disease is marked by a reduction of neural stem cells in the hippocampus dentate gyrus. While increased neural stem cells often correlate with better learning and memory, neurogenesis alone does not always preserve these processes, indicating that other mechanisms involving neural stem cells support memory. It has been shown that intracerebroventricular delivery of neural stem cell-derived small extracellular vesicles in wild-type mice reduces cognitive decline and toxic oligomer binding to synapses. We hypothesize that adequate neural stem cell numbers support neural stem cell-derived small extracellular vesicles protection of synapses against Alzheimer's disease toxic oligomers. Here, we show that elements of immune response in the central nervous system, particularly microglia, may contribute to this protective effect. Specifically, fluorescent-labeled small extracellular vesicles injected into wildtype mice brains were taken up by microglia, with only neural stem cell-derived small extracellular vesicles causing increased microglial activation, indicated by CD68 immunostaining. RNA-sequencing data showed selective activation of immune pathways in microglia by neural stem cell-derived small extracellular vesicles, leading to greater activation and higher Tau uptake 24 hours post-neural stem cell-derived small extracellular vesicle administration. Single-nuclei RNAsequencing of hippocampal microglia gene revealed modulation related to lysosomal activity, supporting neural stem cell-derived small extracellular vesicleinduced neuroprotection via microglia. This study uncovers a novel mechanism through which neural stem cell-derived small extracellular vesicles enhance microglial activity and provide neuroprotection in the hippocampus. Our data demonstrates that neural stem cell-derived small extracellular vesicle uptake by microglia leads to increased microglial activation and improved uptake of Tau oligomers by microglia, suggesting that neural stem cell-derived small extracellular vesicles may prime microglia for a more effective immune response. These results support the hypothesis that neural stem cell-derived small extracellular vesicle-induced modulation of microglial function is crucial for preserving neuronal integrity and mitigating neurodegenerative processes. By elucidating the interactions between neural stem cell-derived small extracellular vesicles and microglia, our study opens new avenues for developing therapeutic strategies aimed at boosting microglial function and addressing neurodegenerative diseases such as Alzheimer's disease.
Additional Links: PMID-41307250
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307250,
year = {2025},
author = {Saieva, S and Scaduto, P and Fracassi, A and Guptarak, J and Zhang, WR and Johnson, K and Jupiter, DC and Marcatti, M and Zolochevska, O and Taglialatela, G and Micci, MA},
title = {Hippocampal neural stem cell-derived extracellular vesicles modulate microglia to promote resilience against tau oligomers.},
journal = {Neural regeneration research},
volume = {},
number = {},
pages = {},
doi = {10.4103/NRR.NRR-D-25-00195},
pmid = {41307250},
issn = {1673-5374},
abstract = {Neural stem cells and adult hippocampal neurogenesis modulate synaptic plasticity and cognitive function. Neural stem cells secrete extracellular vesicles - microvesicles carrying biomolecular cargos - that modulate the function of other cells and contribute to homeostasis and plasticity in the central nervous system. Alzheimer's disease is marked by a reduction of neural stem cells in the hippocampus dentate gyrus. While increased neural stem cells often correlate with better learning and memory, neurogenesis alone does not always preserve these processes, indicating that other mechanisms involving neural stem cells support memory. It has been shown that intracerebroventricular delivery of neural stem cell-derived small extracellular vesicles in wild-type mice reduces cognitive decline and toxic oligomer binding to synapses. We hypothesize that adequate neural stem cell numbers support neural stem cell-derived small extracellular vesicles protection of synapses against Alzheimer's disease toxic oligomers. Here, we show that elements of immune response in the central nervous system, particularly microglia, may contribute to this protective effect. Specifically, fluorescent-labeled small extracellular vesicles injected into wildtype mice brains were taken up by microglia, with only neural stem cell-derived small extracellular vesicles causing increased microglial activation, indicated by CD68 immunostaining. RNA-sequencing data showed selective activation of immune pathways in microglia by neural stem cell-derived small extracellular vesicles, leading to greater activation and higher Tau uptake 24 hours post-neural stem cell-derived small extracellular vesicle administration. Single-nuclei RNAsequencing of hippocampal microglia gene revealed modulation related to lysosomal activity, supporting neural stem cell-derived small extracellular vesicleinduced neuroprotection via microglia. This study uncovers a novel mechanism through which neural stem cell-derived small extracellular vesicles enhance microglial activity and provide neuroprotection in the hippocampus. Our data demonstrates that neural stem cell-derived small extracellular vesicle uptake by microglia leads to increased microglial activation and improved uptake of Tau oligomers by microglia, suggesting that neural stem cell-derived small extracellular vesicles may prime microglia for a more effective immune response. These results support the hypothesis that neural stem cell-derived small extracellular vesicle-induced modulation of microglial function is crucial for preserving neuronal integrity and mitigating neurodegenerative processes. By elucidating the interactions between neural stem cell-derived small extracellular vesicles and microglia, our study opens new avenues for developing therapeutic strategies aimed at boosting microglial function and addressing neurodegenerative diseases such as Alzheimer's disease.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Brain and retina in Alzheimer's disease: Pathological intersections and estimates from imaging.
Alzheimer's & dementia : the journal of the Alzheimer's Association, 21(11):e70884.
Recent studies have highlighted retinal optical coherence tomography (OCT) imaging as a promising biomarker for the early detection of Alzheimer's disease (AD). This review connects AD brain pathology - particularly amyloid beta (Aβ), tau, and vascular changes - with corresponding retinal changes. Evidence suggests that abnormal Aβ and tau deposits in the retina may reflect brain pathology, though their formation mechanisms remain unclear. Retinal vascular changes may also mirror brain pathology, with recent data emerging on other co-pathologies. Retinal thickness changes, especially in acetylcholine-producing layers, can differentiate AD from controls, although not in early AD; however, emerging high-resolution OCT techniques may enhance early detection. We find that correlations between retinal thickness and brain structures are often weak, and retinal vascular imaging shows promise in estimating cerebrovascular disease markers from retinal vascular changes. Novel imaging modalities (e.g., hyperspectral imaging) for detecting retinal Aβ deposits may improve early AD screening when combined with other biomarkers. HIGHLIGHTS: Retinal Aβ/tau is equivocal; peripheral retinal p-tau shows diagnostic promise. OCT retinal/choroid thickness diagnostic/prognostic AUC is small to medium. Hyperspectral imaging and electroretinography may aid early diagnosis. OCTA may differentiate MCI from controls, but preclinical studies are needed. The added value of retinal biomarkers for risk stratification remains uncertain.
Additional Links: PMID-41307192
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307192,
year = {2025},
author = {Banihashemi, MA and Krance, SH and Ji, PX and Koo, M and Ottoy, J and Swartz, RH and Kertes, PJ and Hudson, C and Goubran, M and Black, SE},
title = {Brain and retina in Alzheimer's disease: Pathological intersections and estimates from imaging.},
journal = {Alzheimer's & dementia : the journal of the Alzheimer's Association},
volume = {21},
number = {11},
pages = {e70884},
doi = {10.1002/alz.70884},
pmid = {41307192},
issn = {1552-5279},
support = {/CAPMC/CIHR/Canada ; 177881//CIHR Graduate Scholarships Doctoral Award/ ; //Vision Science Research Program Award/ ; //Peterborough K.M. Hunter Graduate Scholarship/ ; //Queen Elizabeth II/Grace Lumsden and Margaret Nicholds Graduate Scholarship in Science & Technology (QEII-GSST)/ ; //Institute of Medical Science/University of Toronto Open Fellowship Award/ ; //SCACE Graduate Fellowship in Alzheimer Research/ ; //Ontario Graduate Scholarship/ ; 24AARF-1242638/ALZ/Alzheimer's Association/United States ; A2024012F//BrightFocus/ ; //Alzheimer Society of Canada/ ; },
mesh = {Humans ; *Alzheimer Disease/pathology/diagnostic imaging ; Tomography, Optical Coherence ; *Retina/pathology/diagnostic imaging ; *Brain/pathology/diagnostic imaging ; Amyloid beta-Peptides/metabolism ; tau Proteins/metabolism ; Biomarkers ; },
abstract = {Recent studies have highlighted retinal optical coherence tomography (OCT) imaging as a promising biomarker for the early detection of Alzheimer's disease (AD). This review connects AD brain pathology - particularly amyloid beta (Aβ), tau, and vascular changes - with corresponding retinal changes. Evidence suggests that abnormal Aβ and tau deposits in the retina may reflect brain pathology, though their formation mechanisms remain unclear. Retinal vascular changes may also mirror brain pathology, with recent data emerging on other co-pathologies. Retinal thickness changes, especially in acetylcholine-producing layers, can differentiate AD from controls, although not in early AD; however, emerging high-resolution OCT techniques may enhance early detection. We find that correlations between retinal thickness and brain structures are often weak, and retinal vascular imaging shows promise in estimating cerebrovascular disease markers from retinal vascular changes. Novel imaging modalities (e.g., hyperspectral imaging) for detecting retinal Aβ deposits may improve early AD screening when combined with other biomarkers. HIGHLIGHTS: Retinal Aβ/tau is equivocal; peripheral retinal p-tau shows diagnostic promise. OCT retinal/choroid thickness diagnostic/prognostic AUC is small to medium. Hyperspectral imaging and electroretinography may aid early diagnosis. OCTA may differentiate MCI from controls, but preclinical studies are needed. The added value of retinal biomarkers for risk stratification remains uncertain.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alzheimer Disease/pathology/diagnostic imaging
Tomography, Optical Coherence
*Retina/pathology/diagnostic imaging
*Brain/pathology/diagnostic imaging
Amyloid beta-Peptides/metabolism
tau Proteins/metabolism
Biomarkers
RevDate: 2025-11-27
CmpDate: 2025-11-27
Chemogenetic manipulation of learning-tagged neurons is sufficient to rescue progressive memory deficits in a mouse model of Alzheimer's disease.
Alzheimer's & dementia : the journal of the Alzheimer's Association, 21(11):e70953.
INTRODUCTION: Alzheimer's disease (AD) primarily affects episodic memory, which relies on the medial temporal lobe, including the hippocampus and lateral entorhinal cortex (LEC). However, it remains unclear whether memory deficits in AD reflect disrupted encoding of new experiences or impaired retrieval of previously stored information.
METHODS: APPJ20 transgenic mice were used to investigate memory deficits. Neuronal populations activated during the learning phase of associative and non-associative tasks were tagged to express the excitatory chemogenetic receptor hM3Dq. Chemogenetic activation of these tagged neurons was performed during the recall phase of the tasks.
RESULTS: Chemogenetic reactivation of LEC or dentate gyrus (DG) learning-tagged neurons rescued memory performance in associative and non-associative tasks, respectively. Neuronal activation, assessed using c-Fos as a marker, revealed a specific deficit in the reactivation of neurons recruited during learning.
DISCUSSION: Chemogenetic reactivation of neuronal ensembles in the LEC and DG restored memory performance, suggesting that memory deficits in APPJ20 mice are associated with a failure in the endogenous reactivation of learning-relevant neurons.
HIGHLIGHTS: APPJ20 mice exhibited early entorhinal synaptic dysfunction and impaired episodic-like memory retrieval. At a later stage, hippocampal synaptic function became impaired, leading to altered non-associative memory performance. The analysis of neuronal activation using c-Fos revealed a specific impairment of the subpopulation recruited during memory encoding. Chemogenetic reactivation of LEC learning-tagged neurons rescued associative memory performance in 2-month-old APPJ20 mice, while promoting dendritic spine maturation and stabilization in LEC neurons. Chemogenetic reactivation of DG learning-tagged neurons in 6-month-old APPJ20 mice restored non-associative memory retrieval. This study supports the hypothesis that during AD progression, memory is encoded but not accessible through natural cues alone.
Additional Links: PMID-41307180
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307180,
year = {2025},
author = {Guglielmo, S and Di Nardo, N and Scantamburlo, M and Mainardi, M and van den Oever, MC and Origlia, N},
title = {Chemogenetic manipulation of learning-tagged neurons is sufficient to rescue progressive memory deficits in a mouse model of Alzheimer's disease.},
journal = {Alzheimer's & dementia : the journal of the Alzheimer's Association},
volume = {21},
number = {11},
pages = {e70953},
doi = {10.1002/alz.70953},
pmid = {41307180},
issn = {1552-5279},
support = {ECS_00000017//Ministry of University and Research PNRR/ ; DSB.AD005.225//CNR/ ; },
mesh = {Animals ; *Alzheimer Disease/genetics/complications ; Mice, Transgenic ; *Neurons/metabolism/physiology ; Disease Models, Animal ; Mice ; *Memory Disorders/etiology ; Entorhinal Cortex/metabolism ; Dentate Gyrus ; Proto-Oncogene Proteins c-fos/metabolism ; Amyloid beta-Protein Precursor/genetics ; Male ; *Learning/physiology ; Chemogenetics ; },
abstract = {INTRODUCTION: Alzheimer's disease (AD) primarily affects episodic memory, which relies on the medial temporal lobe, including the hippocampus and lateral entorhinal cortex (LEC). However, it remains unclear whether memory deficits in AD reflect disrupted encoding of new experiences or impaired retrieval of previously stored information.
METHODS: APPJ20 transgenic mice were used to investigate memory deficits. Neuronal populations activated during the learning phase of associative and non-associative tasks were tagged to express the excitatory chemogenetic receptor hM3Dq. Chemogenetic activation of these tagged neurons was performed during the recall phase of the tasks.
RESULTS: Chemogenetic reactivation of LEC or dentate gyrus (DG) learning-tagged neurons rescued memory performance in associative and non-associative tasks, respectively. Neuronal activation, assessed using c-Fos as a marker, revealed a specific deficit in the reactivation of neurons recruited during learning.
DISCUSSION: Chemogenetic reactivation of neuronal ensembles in the LEC and DG restored memory performance, suggesting that memory deficits in APPJ20 mice are associated with a failure in the endogenous reactivation of learning-relevant neurons.
HIGHLIGHTS: APPJ20 mice exhibited early entorhinal synaptic dysfunction and impaired episodic-like memory retrieval. At a later stage, hippocampal synaptic function became impaired, leading to altered non-associative memory performance. The analysis of neuronal activation using c-Fos revealed a specific impairment of the subpopulation recruited during memory encoding. Chemogenetic reactivation of LEC learning-tagged neurons rescued associative memory performance in 2-month-old APPJ20 mice, while promoting dendritic spine maturation and stabilization in LEC neurons. Chemogenetic reactivation of DG learning-tagged neurons in 6-month-old APPJ20 mice restored non-associative memory retrieval. This study supports the hypothesis that during AD progression, memory is encoded but not accessible through natural cues alone.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Alzheimer Disease/genetics/complications
Mice, Transgenic
*Neurons/metabolism/physiology
Disease Models, Animal
Mice
*Memory Disorders/etiology
Entorhinal Cortex/metabolism
Dentate Gyrus
Proto-Oncogene Proteins c-fos/metabolism
Amyloid beta-Protein Precursor/genetics
Male
*Learning/physiology
Chemogenetics
RevDate: 2025-11-27
CmpDate: 2025-11-27
Polygenic scores of Core 1 Alzheimer's disease biomarkers predict early cognitive and pathological change.
Alzheimer's & dementia : the journal of the Alzheimer's Association, 21(11):e70937.
INTRODUCTION: Core 1 biomarkers, such as amyloid positron emission tomography, capture the earliest biological changes leading to Alzheimer's disease (AD). While APOE is a major genetic factor, the contribution of other variants to Core 1 biomarkers remains unclear. The goal of this study was to determine whether genetic regulators of Core 1 biomarker levels predicted AD pathology better than genetic regulators of clinical AD.
METHODS: Among 955 non-Hispanic White individuals, polygenic scores (PGSs) were built using genome-wide association studies (GWASs) of amyloid PET, plasma tau phosphorylated at threonine 181 (p-tau181), cerebrospinal fluid (CSF) p-tau181, and clinical AD. Hispanic-specific PGSs were constructed in 515 individuals using plasma p-tau181 and clinical AD GWASs. Baseline and longitudinal associations with plasma biomarkers and cognition were assessed, and replication was conducted in separate cohorts.
RESULTS: The Core 1 biomarker PGSs predicted AD pathology and associated cognitive performance better than the AD PGSs in both populations.
DISCUSSION: The Core 1 PGSs show improved predictive value for AD-related plasma biomarkers and early cognitive changes.
HIGHLIGHTS: APOE ε4 explained more variance in plasma p-tau217 than in plasma p-tau181. PGSs based on Core 1 biomarkers outperformed AD PGSs in predicting plasma biomarkers and cognitive decline among asymptomatic individuals in non-Hispanic White and Hispanic individuals. However, the improvement in predictive power was modest and may vary by age. While the variance in p-tau181 and p-tau217 explained by individual Core 1 PGSs remains limited, the distinct genetic signals captured by the best-performing PGSs across different Core 1 biomarkers may provide an opportunity for developing an integrative Core 1 PGS that more effectively predicts plasma p-tau181 and p-tau217 levels than AD-based PGS.
Additional Links: PMID-41307156
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307156,
year = {2025},
author = {Xu, Y and Qiao, M and Gunasekaran, TI and Gu, Y and Reyes-Dumeyer, D and Piriz, A and Sanchez, D and Soriano, B and Franco, Y and Coronado, ZD and Recio, P and Mejia, DR and Medrano, M and Lantigua, RA and Honig, L and Wilson, R and Manly, JJ and Brickman, AM and Engelman, CD and Johnson, S and Asthana, S and Vardarajan, B and Mayeux, R},
title = {Polygenic scores of Core 1 Alzheimer's disease biomarkers predict early cognitive and pathological change.},
journal = {Alzheimer's & dementia : the journal of the Alzheimer's Association},
volume = {21},
number = {11},
pages = {e70937},
doi = {10.1002/alz.70937},
pmid = {41307156},
issn = {1552-5279},
support = {R01 AG067501/NH/NIH HHS/United States ; R01 AG072474/NH/NIH HHS/United States ; RF1 AG066107/NH/NIH HHS/United States ; UL1TR001873/TR/NCATS NIH HHS/United States ; R01 AG054047/AG/NIA NIH HHS/United States ; RF1 AG054047/AG/NIA NIH HHS/United States ; R01AG027161/AG/NIA NIH HHS/United States ; P30 AG062715/AG/NIA NIH HHS/United States ; },
mesh = {Humans ; *Alzheimer Disease/genetics/pathology ; *Biomarkers/cerebrospinal fluid/blood ; Male ; Female ; *tau Proteins/cerebrospinal fluid/blood ; Aged ; Genome-Wide Association Study ; Positron-Emission Tomography ; *Multifactorial Inheritance/genetics ; Middle Aged ; Cognition ; Aged, 80 and over ; White People/genetics ; },
abstract = {INTRODUCTION: Core 1 biomarkers, such as amyloid positron emission tomography, capture the earliest biological changes leading to Alzheimer's disease (AD). While APOE is a major genetic factor, the contribution of other variants to Core 1 biomarkers remains unclear. The goal of this study was to determine whether genetic regulators of Core 1 biomarker levels predicted AD pathology better than genetic regulators of clinical AD.
METHODS: Among 955 non-Hispanic White individuals, polygenic scores (PGSs) were built using genome-wide association studies (GWASs) of amyloid PET, plasma tau phosphorylated at threonine 181 (p-tau181), cerebrospinal fluid (CSF) p-tau181, and clinical AD. Hispanic-specific PGSs were constructed in 515 individuals using plasma p-tau181 and clinical AD GWASs. Baseline and longitudinal associations with plasma biomarkers and cognition were assessed, and replication was conducted in separate cohorts.
RESULTS: The Core 1 biomarker PGSs predicted AD pathology and associated cognitive performance better than the AD PGSs in both populations.
DISCUSSION: The Core 1 PGSs show improved predictive value for AD-related plasma biomarkers and early cognitive changes.
HIGHLIGHTS: APOE ε4 explained more variance in plasma p-tau217 than in plasma p-tau181. PGSs based on Core 1 biomarkers outperformed AD PGSs in predicting plasma biomarkers and cognitive decline among asymptomatic individuals in non-Hispanic White and Hispanic individuals. However, the improvement in predictive power was modest and may vary by age. While the variance in p-tau181 and p-tau217 explained by individual Core 1 PGSs remains limited, the distinct genetic signals captured by the best-performing PGSs across different Core 1 biomarkers may provide an opportunity for developing an integrative Core 1 PGS that more effectively predicts plasma p-tau181 and p-tau217 levels than AD-based PGS.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alzheimer Disease/genetics/pathology
*Biomarkers/cerebrospinal fluid/blood
Male
Female
*tau Proteins/cerebrospinal fluid/blood
Aged
Genome-Wide Association Study
Positron-Emission Tomography
*Multifactorial Inheritance/genetics
Middle Aged
Cognition
Aged, 80 and over
White People/genetics
RevDate: 2025-11-27
CmpDate: 2025-11-27
Impact of DL-3-n-Butylphthalide on Progression in Alzheimer's Disease: A Retrospective Cohort Analysis.
Neuropsychiatric disease and treatment, 21:2495-2511.
OBJECTIVE: To evaluate the efficacy of DL-3-n-butylphthalide (NBP), a synthetic compound that has shown neuroprotective effects, on cognitive function, psychiatric-behavioral symptoms, and daily activities in patients with Alzheimer's disease (AD).
METHODS: This retrospective cohort study included patients with AD treated with or without NBP. Disease deterioration and decline were defined by changes in Clinical Dementia Rating-Sum of Boxes (CDR-SB) over six months. Multivariate logistic regression, inverse probability of treatment weighting (IPTW) and overlap-weighted propensity score matching (PSM) were used to adjust for confounding.
RESULTS: Totally 100 were included in this study, with 39 classified as the NBP group and 61 as the non-NBP group. NBP was associated with lower odds of deterioration (adjusted odds ratio [OR] = 0.19, 95% confidence interval [CI]: 0.04-0.88, p = 0.034) and decline (adjusted OR = 0.10, 95% CI: 0.03-0.49, p = 0.001). In IPTW and PSM analyses, deterioration occurred in 4.31% vs 22.10% and 4.06% vs 24.27%, and decline in 4.31% vs 39.38% and 4.06% vs 44.28% for the NBP and non-NBP groups, respectively.
CONCLUSION: NBP was associated with reduced risks of clinical worsening and helped preserve cognitive and behavioral functions in patients with AD. These results highlight the potential of NBP as a promising therapeutic option in AD management. Future randomized controlled trials are necessary to validate these findings and assess the long-term efficacy of NBP in clinical settings.
SIGNIFICANCE: This real-world study suggests that NBP may slow disease progression and preserve cognitive and behavioral function in AD.
Additional Links: PMID-41307069
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41307069,
year = {2025},
author = {Zhang, Y and Qiu, J and Shang, Y and Zhao, X and Yang, S and Chen, Y and Dai, S and Ai, M and Huang, W and Zhang, J and Liu, X},
title = {Impact of DL-3-n-Butylphthalide on Progression in Alzheimer's Disease: A Retrospective Cohort Analysis.},
journal = {Neuropsychiatric disease and treatment},
volume = {21},
number = {},
pages = {2495-2511},
pmid = {41307069},
issn = {1176-6328},
abstract = {OBJECTIVE: To evaluate the efficacy of DL-3-n-butylphthalide (NBP), a synthetic compound that has shown neuroprotective effects, on cognitive function, psychiatric-behavioral symptoms, and daily activities in patients with Alzheimer's disease (AD).
METHODS: This retrospective cohort study included patients with AD treated with or without NBP. Disease deterioration and decline were defined by changes in Clinical Dementia Rating-Sum of Boxes (CDR-SB) over six months. Multivariate logistic regression, inverse probability of treatment weighting (IPTW) and overlap-weighted propensity score matching (PSM) were used to adjust for confounding.
RESULTS: Totally 100 were included in this study, with 39 classified as the NBP group and 61 as the non-NBP group. NBP was associated with lower odds of deterioration (adjusted odds ratio [OR] = 0.19, 95% confidence interval [CI]: 0.04-0.88, p = 0.034) and decline (adjusted OR = 0.10, 95% CI: 0.03-0.49, p = 0.001). In IPTW and PSM analyses, deterioration occurred in 4.31% vs 22.10% and 4.06% vs 24.27%, and decline in 4.31% vs 39.38% and 4.06% vs 44.28% for the NBP and non-NBP groups, respectively.
CONCLUSION: NBP was associated with reduced risks of clinical worsening and helped preserve cognitive and behavioral functions in patients with AD. These results highlight the potential of NBP as a promising therapeutic option in AD management. Future randomized controlled trials are necessary to validate these findings and assess the long-term efficacy of NBP in clinical settings.
SIGNIFICANCE: This real-world study suggests that NBP may slow disease progression and preserve cognitive and behavioral function in AD.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Genetic knockdown of DYRK1A attenuates cognitive impairment, Aβ pathology, tauopathy and neuroinflammatory responses in mouse models of AD.
Frontiers in immunology, 16:1661791.
INTRODUCTION: Dual specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is associated with the pathoprogression of neurodevelopmental and neurodegenerative disorders. However, the effects of direct genetic manipulation of DYRK1A in the brain on cognitive function, neuroinflammation and Alzheimer's disease (AD) pathology and underlying molecular mechanisms have not been fully investigated.
METHODS: To determine whether overexpressing or knocking down DYRK1A expression directly in the brain affects cognitive function, neuroinflammation and AD pathology, adeno-associated viruses (AAVs) were injected into the hippocampus of wild-type (WT), 5xFAD, and PS19 mice. Then, cognitive function was assessed via Y-maze and novel object recognition (NOR) tests, and neuroinflammatory responses and AD pathologies were analyzed by real-time PCR, Western blotting, immunofluorescence staining, AD-associated protein activity assays and ELISA.
RESULTS AND DISCUSSION: In WT mice, hippocampal DYRK1A overexpression significantly reduced short-term spatial/recognition memory and SynGAP expression while increasing p-P38 levels. Conversely, in amyloid-beta (Aβ)-overexpressing 5xFAD mice, hippocampal DYRK1A knockdown improved short-term spatial/recognition memory and significantly increased CaMKIIα and CREB phosphorylation. Moreover, hippocampal DYRK1A knockdown in 5xFAD mice significantly suppressed mRNA levels of proinflammatory cytokines and markers of AD-associated reactive astrocytes (RAs), disease-associated microglia (DAMs), and RA-DAM interactions. However, hippocampal DYRK1A overexpression in 5xFAD mice increased mRNA levels of the proinflammatory cytokine IL-1β, RA markers and the microglial marker Iba-1. Interestingly, hippocampal DYRK1A knockdown in 5xFAD mice significantly increased levels of the anti-oxidative/inflammatory molecule HO-1 without altering p-STAT3/p-NF-κB levels. By contrast, hippocampal DYRK1A overexpression in 5xFAD mice enhanced STAT3/NF-κB phosphorylation but did not affect ROS levels. Importantly, hippocampal DYRK1A knockdown in 5xFAD mice significantly reduced Aβ plaque number, soluble Aβ40 levels, and soluble/insoluble Aβ42 levels by suppressing β-secretase BACE1 activity but not tau hyperphosphorylation. Finally, hippocampal DYRK1A knockdown in PS19 mice [a model of AD that overexpresses human mutant tau (P301S)] selectively decreased insoluble tau hyperphosphorylation at Ser396 and Ser404 and alleviated proinflammatory responses/glial-associated neuroinflammatory dynamics. Taken together, our data indicate that DYRK1A modulates cognitive function, neuroinflammation, and AD pathology (Aβ and tauopathy) in mouse models of AD and/or WT mice and support DYRK1A as a potential therapeutic target for AD.
Additional Links: PMID-41306975
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306975,
year = {2025},
author = {Lee, HJ and Kang, S and Lee, YJ and Oh, S and Joo, B and Hwang, JW and Kim, J and Kim, TE and Jung, TM and Kim, YJ and Jang, JY and Song, JH and Koo, JW and Hoe, HS},
title = {Genetic knockdown of DYRK1A attenuates cognitive impairment, Aβ pathology, tauopathy and neuroinflammatory responses in mouse models of AD.},
journal = {Frontiers in immunology},
volume = {16},
number = {},
pages = {1661791},
pmid = {41306975},
issn = {1664-3224},
mesh = {Animals ; Dyrk Kinases ; *Protein-Tyrosine Kinases/genetics/metabolism ; Mice ; Disease Models, Animal ; *Alzheimer Disease/genetics/pathology/metabolism ; *Protein Serine-Threonine Kinases/genetics/metabolism ; *Amyloid beta-Peptides/metabolism ; Mice, Transgenic ; *Cognitive Dysfunction/genetics/metabolism/pathology ; *Tauopathies/genetics/pathology/metabolism ; Hippocampus/metabolism/pathology ; *Neuroinflammatory Diseases/genetics/pathology/metabolism ; Male ; Gene Knockdown Techniques ; Humans ; Mice, Inbred C57BL ; },
abstract = {INTRODUCTION: Dual specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is associated with the pathoprogression of neurodevelopmental and neurodegenerative disorders. However, the effects of direct genetic manipulation of DYRK1A in the brain on cognitive function, neuroinflammation and Alzheimer's disease (AD) pathology and underlying molecular mechanisms have not been fully investigated.
METHODS: To determine whether overexpressing or knocking down DYRK1A expression directly in the brain affects cognitive function, neuroinflammation and AD pathology, adeno-associated viruses (AAVs) were injected into the hippocampus of wild-type (WT), 5xFAD, and PS19 mice. Then, cognitive function was assessed via Y-maze and novel object recognition (NOR) tests, and neuroinflammatory responses and AD pathologies were analyzed by real-time PCR, Western blotting, immunofluorescence staining, AD-associated protein activity assays and ELISA.
RESULTS AND DISCUSSION: In WT mice, hippocampal DYRK1A overexpression significantly reduced short-term spatial/recognition memory and SynGAP expression while increasing p-P38 levels. Conversely, in amyloid-beta (Aβ)-overexpressing 5xFAD mice, hippocampal DYRK1A knockdown improved short-term spatial/recognition memory and significantly increased CaMKIIα and CREB phosphorylation. Moreover, hippocampal DYRK1A knockdown in 5xFAD mice significantly suppressed mRNA levels of proinflammatory cytokines and markers of AD-associated reactive astrocytes (RAs), disease-associated microglia (DAMs), and RA-DAM interactions. However, hippocampal DYRK1A overexpression in 5xFAD mice increased mRNA levels of the proinflammatory cytokine IL-1β, RA markers and the microglial marker Iba-1. Interestingly, hippocampal DYRK1A knockdown in 5xFAD mice significantly increased levels of the anti-oxidative/inflammatory molecule HO-1 without altering p-STAT3/p-NF-κB levels. By contrast, hippocampal DYRK1A overexpression in 5xFAD mice enhanced STAT3/NF-κB phosphorylation but did not affect ROS levels. Importantly, hippocampal DYRK1A knockdown in 5xFAD mice significantly reduced Aβ plaque number, soluble Aβ40 levels, and soluble/insoluble Aβ42 levels by suppressing β-secretase BACE1 activity but not tau hyperphosphorylation. Finally, hippocampal DYRK1A knockdown in PS19 mice [a model of AD that overexpresses human mutant tau (P301S)] selectively decreased insoluble tau hyperphosphorylation at Ser396 and Ser404 and alleviated proinflammatory responses/glial-associated neuroinflammatory dynamics. Taken together, our data indicate that DYRK1A modulates cognitive function, neuroinflammation, and AD pathology (Aβ and tauopathy) in mouse models of AD and/or WT mice and support DYRK1A as a potential therapeutic target for AD.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
Dyrk Kinases
*Protein-Tyrosine Kinases/genetics/metabolism
Mice
Disease Models, Animal
*Alzheimer Disease/genetics/pathology/metabolism
*Protein Serine-Threonine Kinases/genetics/metabolism
*Amyloid beta-Peptides/metabolism
Mice, Transgenic
*Cognitive Dysfunction/genetics/metabolism/pathology
*Tauopathies/genetics/pathology/metabolism
Hippocampus/metabolism/pathology
*Neuroinflammatory Diseases/genetics/pathology/metabolism
Male
Gene Knockdown Techniques
Humans
Mice, Inbred C57BL
RevDate: 2025-11-27
CmpDate: 2025-11-27
Leveraging recent advances in plasma biomarkers to optimize early proof of concept trials in Alzheimer's disease.
Alzheimer's & dementia (New York, N. Y.), 11(4):e70183.
INTRODUCTION: The importance of biomarkers as a primary outcome or as supportive evidence of clinical effect is rising as the field shifts toward disease-modifying treatments and earlier intervention, because they have lower variability and can indicate disease progression earlier than clinical outcomes. This study assessed the performance of plasma pTau 181 and 217 as a predictive biomarker and potential primary endpoint in early-phase Alzheimer's disease (AD) trials.
METHODS: Summary data from recent monoclonal antibody (mAb) trials including plasma pTau 181 and 217 were analyzed to evaluate associations between plasma pTau 181/217 and clinical outcomes. The suitability of plasma pTau 181/217 as a surrogate endpoint for internal decision making was assessed using Prentice criteria. Simulations were conducted to explore the statistical power of using plasma pTau 181/217 as a primary outcome in dose-escalation, proof-of-concept (POC) trial designs. Additional criteria for biomarker validation were applied to simulated data.
RESULTS: A strong group-level correlation (r = 0.781) was observed between treatment effects on plasma pTau 181/217 and Clinical Dementia Rating scale - Sum of Boxes (CDR-SB). Mean change in plasma pTau 181/217 significantly predicted mean change in CDR-SB (p = 0.013). The treatment effect on pTau 181/217 was ∼2.6 times greater than on CDR-SB. Prentice Criteria 1, 2, and 4 were met or reasonably met; Criterion 3 is not applicable in the POC setting.
CONCLUSION: Plasma pTau 181/217 at 6 months shows future promise to reasonably likely predict clinical benefit for drugs that reduce pTau 181/217 levels, supporting its use as a primary endpoint in early-phase trials. With effect sizes similar to those seen with donanemab, adequately powered trials may require as few as 100 participants. Such trials should include prespecified analyses to evaluate individual-level Prentice criteria, and pTau 181/217 results can be used to predict potential Phase 3 clinical outcomes.
HIGHLIGHTS: The group-level correlation between a biomarker treatment effect and clinical endpoint treatment effect is a measurement of the biomarker's ability to predict clinical outcome.The correlation of group level plasma pT217 or pT181 effect size at 6 months with clinical outcome Clinical Dementia Rating scale - Sum of Boxes (CDR-SB) effect size at 12 months was approximately 0.781 with p values of 0.013.Cohen's d effect size of plasma pTau as an outcome was 2.6 times greater than the Cohen's d of CDR-SB, leading to higher power or lower sample sizes.As a primary endpoint, plasma pTau meets or reasonably meets Prentice Criteria 1, 2, and 4, while Criterion 3 was deemed not applicable in the proof-of-concept study setting.
Additional Links: PMID-41306895
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306895,
year = {2025},
author = {Duncan, GB and Dickson, SP and Kaplan, JM and Johnson, SB and Duke, TM and Dayley, CW and Hendrix, SB and Altstiel, LD and Mallinckrodt, CH},
title = {Leveraging recent advances in plasma biomarkers to optimize early proof of concept trials in Alzheimer's disease.},
journal = {Alzheimer's & dementia (New York, N. Y.)},
volume = {11},
number = {4},
pages = {e70183},
pmid = {41306895},
issn = {2352-8737},
abstract = {INTRODUCTION: The importance of biomarkers as a primary outcome or as supportive evidence of clinical effect is rising as the field shifts toward disease-modifying treatments and earlier intervention, because they have lower variability and can indicate disease progression earlier than clinical outcomes. This study assessed the performance of plasma pTau 181 and 217 as a predictive biomarker and potential primary endpoint in early-phase Alzheimer's disease (AD) trials.
METHODS: Summary data from recent monoclonal antibody (mAb) trials including plasma pTau 181 and 217 were analyzed to evaluate associations between plasma pTau 181/217 and clinical outcomes. The suitability of plasma pTau 181/217 as a surrogate endpoint for internal decision making was assessed using Prentice criteria. Simulations were conducted to explore the statistical power of using plasma pTau 181/217 as a primary outcome in dose-escalation, proof-of-concept (POC) trial designs. Additional criteria for biomarker validation were applied to simulated data.
RESULTS: A strong group-level correlation (r = 0.781) was observed between treatment effects on plasma pTau 181/217 and Clinical Dementia Rating scale - Sum of Boxes (CDR-SB). Mean change in plasma pTau 181/217 significantly predicted mean change in CDR-SB (p = 0.013). The treatment effect on pTau 181/217 was ∼2.6 times greater than on CDR-SB. Prentice Criteria 1, 2, and 4 were met or reasonably met; Criterion 3 is not applicable in the POC setting.
CONCLUSION: Plasma pTau 181/217 at 6 months shows future promise to reasonably likely predict clinical benefit for drugs that reduce pTau 181/217 levels, supporting its use as a primary endpoint in early-phase trials. With effect sizes similar to those seen with donanemab, adequately powered trials may require as few as 100 participants. Such trials should include prespecified analyses to evaluate individual-level Prentice criteria, and pTau 181/217 results can be used to predict potential Phase 3 clinical outcomes.
HIGHLIGHTS: The group-level correlation between a biomarker treatment effect and clinical endpoint treatment effect is a measurement of the biomarker's ability to predict clinical outcome.The correlation of group level plasma pT217 or pT181 effect size at 6 months with clinical outcome Clinical Dementia Rating scale - Sum of Boxes (CDR-SB) effect size at 12 months was approximately 0.781 with p values of 0.013.Cohen's d effect size of plasma pTau as an outcome was 2.6 times greater than the Cohen's d of CDR-SB, leading to higher power or lower sample sizes.As a primary endpoint, plasma pTau meets or reasonably meets Prentice Criteria 1, 2, and 4, while Criterion 3 was deemed not applicable in the proof-of-concept study setting.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Differential effects of Huang-Lian-Jie-Du Decoction on Alzheimer's disease and normal rats.
Frontiers in pharmacology, 16:1710919.
INTRODUCTION: Huang-Lian-Jie-Du Decoction (HLJDD), a botanical drug used in traditional medicine, has been used in the management of Alzheimer's disease (AD). However, the mechanisms underlying its preventive effects remain inadequately understood, particularly due to the absence of metabolomic studies examining alterations in serum and cerebrospinal fluid (CSF) metabolites. Moreover, the potential toxicities and side effects of HLJDD necessitate further pharmacological investigation. This study aims to explore the differential effects of HLJDD on AD model rats and healthy controls through a metabolomics approach and uncover the underlying mechanisms based on changes in serum and CSF metabolites. The findings are expected to provide a scientific foundation for enhancing the clinical safety and rational use of HLJDD.
METHODS: The composition of HLJDD was characterized by UPLC-Q-Exactive Orbitrap HRMS. Aβ1-42-induced SD rats served as the AD animal model. Rats in the sham + HLJDD and Aβ1-42 + HLJDD groups (0.604 g/kg freeze-dried powder) were treated with HLJDD via gavage for 28 days. Nissl staining was performed to assess hippocampal neuronal changes, while H&E staining was used to evaluate histopathological alterations in the brain, liver, kidneys, stomach, large intestine, and small intestine. Aβ expression was determined using IHC and ELISA, and inflammatory levels in both peripheral and central systems were quantified by ELISA. MMP-2 and MMP-9 expression were analyzed through IHC. LC-MS was employed to investigate metabolic variations in serum and CSF.
RESULTS: HLJDD reduced Aβ deposition in Alzheimer's disease rats, enhanced neuronal survival, reduced inflammation, preserved blood-brain barrier (BBB) integrity, and alleviated damage to the brain, kidneys, and stomach. These therapeutic effects were associated with the arginine biosynthesis pathway and ferroptosis. In contrast, HLJDD induced peripheral and central inflammation, impaired neuronal function, compromised BBB integrity, and caused damage to the liver, kidneys, and large intestine in normal rats. These adverse effects were linked to disruptions in aminobenzoate degradation and nucleotide metabolism.
CONCLUSION: HLJDD may alleviate Aβ-induced damage repair in Alzheimer's disease rats, but it also induces varying degrees of toxicity in normal rats.
Additional Links: PMID-41306777
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306777,
year = {2025},
author = {Xu, M and Zhu, Y and Chen, J and Zhong, F and Wang, R and Li, J and Qiao, M and Fan, Y and Ren, P and Chen, M and Qin, J and Wu, W},
title = {Differential effects of Huang-Lian-Jie-Du Decoction on Alzheimer's disease and normal rats.},
journal = {Frontiers in pharmacology},
volume = {16},
number = {},
pages = {1710919},
pmid = {41306777},
issn = {1663-9812},
abstract = {INTRODUCTION: Huang-Lian-Jie-Du Decoction (HLJDD), a botanical drug used in traditional medicine, has been used in the management of Alzheimer's disease (AD). However, the mechanisms underlying its preventive effects remain inadequately understood, particularly due to the absence of metabolomic studies examining alterations in serum and cerebrospinal fluid (CSF) metabolites. Moreover, the potential toxicities and side effects of HLJDD necessitate further pharmacological investigation. This study aims to explore the differential effects of HLJDD on AD model rats and healthy controls through a metabolomics approach and uncover the underlying mechanisms based on changes in serum and CSF metabolites. The findings are expected to provide a scientific foundation for enhancing the clinical safety and rational use of HLJDD.
METHODS: The composition of HLJDD was characterized by UPLC-Q-Exactive Orbitrap HRMS. Aβ1-42-induced SD rats served as the AD animal model. Rats in the sham + HLJDD and Aβ1-42 + HLJDD groups (0.604 g/kg freeze-dried powder) were treated with HLJDD via gavage for 28 days. Nissl staining was performed to assess hippocampal neuronal changes, while H&E staining was used to evaluate histopathological alterations in the brain, liver, kidneys, stomach, large intestine, and small intestine. Aβ expression was determined using IHC and ELISA, and inflammatory levels in both peripheral and central systems were quantified by ELISA. MMP-2 and MMP-9 expression were analyzed through IHC. LC-MS was employed to investigate metabolic variations in serum and CSF.
RESULTS: HLJDD reduced Aβ deposition in Alzheimer's disease rats, enhanced neuronal survival, reduced inflammation, preserved blood-brain barrier (BBB) integrity, and alleviated damage to the brain, kidneys, and stomach. These therapeutic effects were associated with the arginine biosynthesis pathway and ferroptosis. In contrast, HLJDD induced peripheral and central inflammation, impaired neuronal function, compromised BBB integrity, and caused damage to the liver, kidneys, and large intestine in normal rats. These adverse effects were linked to disruptions in aminobenzoate degradation and nucleotide metabolism.
CONCLUSION: HLJDD may alleviate Aβ-induced damage repair in Alzheimer's disease rats, but it also induces varying degrees of toxicity in normal rats.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Riboflavin-sensitized photoinduced degradation of donepezil hydrochloride: kinetic and computational insights for pharmaceutical formulation.
RSC advances, 15(53):45799-45821.
Donepezil (DPZ) photosensitized degradation by riboflavin (RF) at pH 2.0-12.0 has been carried out in aerobic and anaerobic conditions under UV and visible irradiation to give an idea to pharmaceutical formulators to develop a stable co-formulation to administer to elderly patients with neurodegenerative diseases (Alzheimer's disease, Parkinson's disease). The photolysis rate constants (k obs) in aerobic and anaerobic conditions in the presence of RF (0.1-0.5 × 10[-4] M) range from 0.25-11.5 and 0.025-1.120 × 10[-2] min[-1], respectively. RF catalyzes the photodegradation of DPZ, and the second-order (k 2) constants in aerobic and anaerobic conditions range from 0.32 to 8.22 and 0.032 to 0.805 × 10[-2] M[-1] min[-1], respectively, indicating that with an increase in the concentrations of RF, the rate of photolysis of DPZ also increases. The k-pH profile shows a bell-shaped curve from pH 2.0 to 4.0 and from pH 5.0 to 12.0, forming a sigmoid curve. The two-component spectrometric and HPLC green methods have been developed and validated for estimating DPZ and RF in pure and degraded solutions. Additionally, computational studies have been conducted to assess the formation of the ground-state complex, binding affinity, and molecular interactions between DPZ and RF.
Additional Links: PMID-41306676
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306676,
year = {2025},
author = {Khan, T and Anwar, Z and Altaf, R and Awan, A and Noreen, A and Usmani, M and Ejaz, MA and Khan, A and Ahmed, S and Sheraz, MA and Ali, T},
title = {Riboflavin-sensitized photoinduced degradation of donepezil hydrochloride: kinetic and computational insights for pharmaceutical formulation.},
journal = {RSC advances},
volume = {15},
number = {53},
pages = {45799-45821},
pmid = {41306676},
issn = {2046-2069},
abstract = {Donepezil (DPZ) photosensitized degradation by riboflavin (RF) at pH 2.0-12.0 has been carried out in aerobic and anaerobic conditions under UV and visible irradiation to give an idea to pharmaceutical formulators to develop a stable co-formulation to administer to elderly patients with neurodegenerative diseases (Alzheimer's disease, Parkinson's disease). The photolysis rate constants (k obs) in aerobic and anaerobic conditions in the presence of RF (0.1-0.5 × 10[-4] M) range from 0.25-11.5 and 0.025-1.120 × 10[-2] min[-1], respectively. RF catalyzes the photodegradation of DPZ, and the second-order (k 2) constants in aerobic and anaerobic conditions range from 0.32 to 8.22 and 0.032 to 0.805 × 10[-2] M[-1] min[-1], respectively, indicating that with an increase in the concentrations of RF, the rate of photolysis of DPZ also increases. The k-pH profile shows a bell-shaped curve from pH 2.0 to 4.0 and from pH 5.0 to 12.0, forming a sigmoid curve. The two-component spectrometric and HPLC green methods have been developed and validated for estimating DPZ and RF in pure and degraded solutions. Additionally, computational studies have been conducted to assess the formation of the ground-state complex, binding affinity, and molecular interactions between DPZ and RF.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Anti-Amyloid Therapies for Alzheimer's Disease: Not the Way Forward.
Canadian geriatrics journal : CGJ, 28(4):400-402.
Additional Links: PMID-41306639
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306639,
year = {2025},
author = {Bocti, C and Bergman, H},
title = {Anti-Amyloid Therapies for Alzheimer's Disease: Not the Way Forward.},
journal = {Canadian geriatrics journal : CGJ},
volume = {28},
number = {4},
pages = {400-402},
pmid = {41306639},
issn = {1925-8348},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Predicting risk of impending cognitive decline in asymptomatic individuals with early Alzheimer's disease: Insights from cortical diffusion MRI.
Imaging neuroscience (Cambridge, Mass.), 3:.
Neurodegeneration associated with Alzheimer's disease (AD) can start at the microstructural level years before cognitive symptoms; yet, it has been difficult to definitively detect these early changes to plan effective treatments. Using a cortical column-based analysis of high-resolution diffusion magnetic resonance imaging data, we aim to identify early microstructural neurodegeneration in the gray matter across different cortical depths and regions. We compared four groups of participants across a spectrum of cognitive decline (n = 60): cognitively normal amyloid-negative (normal controls), cognitively normal amyloid-positive (stage-1 AD), mild cognitive impairment (MCI), and AD dementia. Our results showed progressive increases in radial diffusivity across these groups in cortical regions associated with AD, and our analysis in individual asymptomatic stage-1 AD participants was able to differentiate such diffusivity increases to predict risk of impending cognitive decline in 2 participants who had cognitive decline and MCI diagnosis at their follow-up clinical examination and 11 participants who did not.
Additional Links: PMID-41306598
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306598,
year = {2025},
author = {Overson, DK and Truong, TK and Petrella, JR and Madden, DJ and Ma, Y and Johnson, KG and Liu, AJ and O'Brien, RJ and Whitson, HE and Song, AW},
title = {Predicting risk of impending cognitive decline in asymptomatic individuals with early Alzheimer's disease: Insights from cortical diffusion MRI.},
journal = {Imaging neuroscience (Cambridge, Mass.)},
volume = {3},
number = {},
pages = {},
pmid = {41306598},
issn = {2837-6056},
abstract = {Neurodegeneration associated with Alzheimer's disease (AD) can start at the microstructural level years before cognitive symptoms; yet, it has been difficult to definitively detect these early changes to plan effective treatments. Using a cortical column-based analysis of high-resolution diffusion magnetic resonance imaging data, we aim to identify early microstructural neurodegeneration in the gray matter across different cortical depths and regions. We compared four groups of participants across a spectrum of cognitive decline (n = 60): cognitively normal amyloid-negative (normal controls), cognitively normal amyloid-positive (stage-1 AD), mild cognitive impairment (MCI), and AD dementia. Our results showed progressive increases in radial diffusivity across these groups in cortical regions associated with AD, and our analysis in individual asymptomatic stage-1 AD participants was able to differentiate such diffusivity increases to predict risk of impending cognitive decline in 2 participants who had cognitive decline and MCI diagnosis at their follow-up clinical examination and 11 participants who did not.},
}
RevDate: 2025-11-27
The Link Between Circadian Disruption and Alzheimer's Disease and Related Dementias: Insights from Peripheral Inflammation.
Current opinion in physiology, 46:.
Circadian rhythms are essential for maintaining physiological homeostasis, influencing biological processes from the sleep-wake cycle to metabolism and immune responses. Disruption of these rhythms is increasingly linked to the pathogenesis of Alzheimer's Disease and Related Dementias (ADRDs), conditions characterized by cognitive decline and neuropsychiatric symptoms through various pathways including increases in inflammation. While many studies link the effects of circadian disruption on neuroinflammation to ADRDs, this review explores the potential link between the circadian disruption of peripheral inflammation and ADRDs. We discuss the evidence of how circadian misalignment can exacerbate neuroinflammation through the activation of the peripheral immune system. We further examine the role of peripheral factors such as insulin dysregulation, melatonin levels, and gut microbiome imbalances in amplifying these peripheral inflammatory responses. These data underscore the significance of circadian regulation in maintaining immune homeostasis, highlighting potential therapeutic avenues for mitigating ADRDs through the restoration of circadian integrity.
Additional Links: PMID-41306540
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306540,
year = {2025},
author = {Cordi, CV and Hurley, JM},
title = {The Link Between Circadian Disruption and Alzheimer's Disease and Related Dementias: Insights from Peripheral Inflammation.},
journal = {Current opinion in physiology},
volume = {46},
number = {},
pages = {},
pmid = {41306540},
issn = {2468-8673},
abstract = {Circadian rhythms are essential for maintaining physiological homeostasis, influencing biological processes from the sleep-wake cycle to metabolism and immune responses. Disruption of these rhythms is increasingly linked to the pathogenesis of Alzheimer's Disease and Related Dementias (ADRDs), conditions characterized by cognitive decline and neuropsychiatric symptoms through various pathways including increases in inflammation. While many studies link the effects of circadian disruption on neuroinflammation to ADRDs, this review explores the potential link between the circadian disruption of peripheral inflammation and ADRDs. We discuss the evidence of how circadian misalignment can exacerbate neuroinflammation through the activation of the peripheral immune system. We further examine the role of peripheral factors such as insulin dysregulation, melatonin levels, and gut microbiome imbalances in amplifying these peripheral inflammatory responses. These data underscore the significance of circadian regulation in maintaining immune homeostasis, highlighting potential therapeutic avenues for mitigating ADRDs through the restoration of circadian integrity.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Expression of dementia biomarkers in Appalachian and non-Appalachian ELVO patients during thrombectomy.
Frontiers in neuroscience, 19:1672803.
BACKGROUND/CONTEXT: Vascular Cognitive Impairment and Dementia (VCID) affects 25-30% of stroke patients and includes cognitive impairments caused by vascular injury, such as post-stroke dementia. Rehabilitation has the potential to improve the quality of life for patients at risk of developing dementia. However, there is currently no reliable method to identify those at risk of dementia after a stroke. Several biomarkers, including ADRD (Alzheimer's disease and related dementias) biomarkers (Ab, tau, NfL, and GFAP) and angiogenic factors (VEGF, Flt-1, Tie-2, PIGF, and FGF) have been associated with the development of dementia.Populations in Appalachia experience a higher incidence of stroke and related mortality compared to other groups. Given the elevated stroke rates in Appalachian communities, this study aims to investigate potential proteomic differences between patients from Appalachian and non-Appalachian counties. The primary goal of the study is to characterize the expression of post-stroke cognitive dementia biomarkers and to explore differences in the proteomic profiles of Appalachian and non-Appalachian populations.
METHODS/APPROACH: Sample Collection: The Blood and Clot Thrombectomy Registry Collaborative (BACTRAC) protocol, established by Fraser and colleagues, introduces a novel method for analyzing stroke by collecting intracranial blood samples from patients undergoing mechanical thrombectomy. During the procedure the thrombus and blood samples from areas distal and proximal to the thrombus are collected and undergo proteomic analysis (Fraser et al.). Additional demographic and clinical information are collected from electronic health records. The control data was obtained from arterial blood collected during diagnostic angiograms from patients with cerebrovascular disease.
DATA ANALYSIS: Propensity score models were used to perform a one-to-one match between stroke and control patients on age, sex, BMI, hypertension, and hyperlipidemia resulting in groups that were balanced on these measured prognostic characteristics. A Wilcoxon rank sum test was then used to assess differences in the 12 ADRD biomarkers.
RESULTS: Compared to the controls, stroke patients had significantly higher levels of GFAP. The control patients had significantly higher levels of AB40, AB42, and VEGFA. In the Appalachian patient population, the control patients also had significantly higher levels of AB40, AB42, and VEGFA. Additionally, the Appalachian stroke patients had higher GFAP. In the non-Appalachian population only GFAP was significantly different between stroke and control groups, with it being elevated in the stroke group.
CONCLUSION: There was a notable difference in the levels of certain ADRD biomarkers between stroke patients and control patients. Specifically, in Appalachian populations, stroke patients showed significant differences in multiple ADRD biomarkers (AB40, AB42, and GFAP) compared to controls, a pattern not seen in non-Appalachian stroke patients, where only GFAP levels increased. This difference in ADRD biomarkers observed in Appalachian stroke patients could be attributed to a combination of socioeconomic and environmental factors unique to the Appalachian region.
Additional Links: PMID-41306424
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306424,
year = {2025},
author = {Anil, N and McLouth, CJ and Hazelwood, HS and Dahlke, E and Frank, JA and Millson, N and Al-Kawaz, M and Harp, JP and Cranford, W and Pahwa, S and Dornbos, D and Fraser, JF and Pennypacker, KR},
title = {Expression of dementia biomarkers in Appalachian and non-Appalachian ELVO patients during thrombectomy.},
journal = {Frontiers in neuroscience},
volume = {19},
number = {},
pages = {1672803},
pmid = {41306424},
issn = {1662-4548},
abstract = {BACKGROUND/CONTEXT: Vascular Cognitive Impairment and Dementia (VCID) affects 25-30% of stroke patients and includes cognitive impairments caused by vascular injury, such as post-stroke dementia. Rehabilitation has the potential to improve the quality of life for patients at risk of developing dementia. However, there is currently no reliable method to identify those at risk of dementia after a stroke. Several biomarkers, including ADRD (Alzheimer's disease and related dementias) biomarkers (Ab, tau, NfL, and GFAP) and angiogenic factors (VEGF, Flt-1, Tie-2, PIGF, and FGF) have been associated with the development of dementia.Populations in Appalachia experience a higher incidence of stroke and related mortality compared to other groups. Given the elevated stroke rates in Appalachian communities, this study aims to investigate potential proteomic differences between patients from Appalachian and non-Appalachian counties. The primary goal of the study is to characterize the expression of post-stroke cognitive dementia biomarkers and to explore differences in the proteomic profiles of Appalachian and non-Appalachian populations.
METHODS/APPROACH: Sample Collection: The Blood and Clot Thrombectomy Registry Collaborative (BACTRAC) protocol, established by Fraser and colleagues, introduces a novel method for analyzing stroke by collecting intracranial blood samples from patients undergoing mechanical thrombectomy. During the procedure the thrombus and blood samples from areas distal and proximal to the thrombus are collected and undergo proteomic analysis (Fraser et al.). Additional demographic and clinical information are collected from electronic health records. The control data was obtained from arterial blood collected during diagnostic angiograms from patients with cerebrovascular disease.
DATA ANALYSIS: Propensity score models were used to perform a one-to-one match between stroke and control patients on age, sex, BMI, hypertension, and hyperlipidemia resulting in groups that were balanced on these measured prognostic characteristics. A Wilcoxon rank sum test was then used to assess differences in the 12 ADRD biomarkers.
RESULTS: Compared to the controls, stroke patients had significantly higher levels of GFAP. The control patients had significantly higher levels of AB40, AB42, and VEGFA. In the Appalachian patient population, the control patients also had significantly higher levels of AB40, AB42, and VEGFA. Additionally, the Appalachian stroke patients had higher GFAP. In the non-Appalachian population only GFAP was significantly different between stroke and control groups, with it being elevated in the stroke group.
CONCLUSION: There was a notable difference in the levels of certain ADRD biomarkers between stroke patients and control patients. Specifically, in Appalachian populations, stroke patients showed significant differences in multiple ADRD biomarkers (AB40, AB42, and GFAP) compared to controls, a pattern not seen in non-Appalachian stroke patients, where only GFAP levels increased. This difference in ADRD biomarkers observed in Appalachian stroke patients could be attributed to a combination of socioeconomic and environmental factors unique to the Appalachian region.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Intracranial self-stimulation mitigates spatial task deficits, modifies miR-146a and miR-495 serum levels and restores hippocampal NRF2 levels in a rat model of sporadic Alzheimer's disease.
Frontiers in aging neuroscience, 17:1671196.
INTRODUCTION: Alzheimer's disease (AD) is the most prevalent neurodegenerative disease associated with aging. While deep brain stimulation (DBS) shows therapeutic promise, the long-term persistence of its effects remains understudied. Expression patterns of circulating miRNAs, proposed diagnostic biomarkers, and their modulation by DBS are still poorly characterized in longitudinal studies. This study investigates the effect of a 13-week prolonged ICSS treatment on spatial memory and serum miRNA expression in a male rat model of sporadic AD (SAD) by intracerebroventricular injection of streptozotocin (STZ).
METHODS: Morris water maze (MWM) tasks were conducted at 1 and 5 months post-STZ. Serum miRNA levels were quantified by qRT-PCR at 29 (Ser0), 73 (Ser1) and 136 (Ser2) days after STZ administration. Corpus callosum thickness and NRF2 protein levels in the hippocampal CA1 region were assessed using Nissl staining and immunohistochemistry, respectively. Target validation of miR-495 was performed via transfection assays in the human neuroblastoma SH-SY5Y cell line.
RESULTS: MFB-ICSS treatment significantly reduced escape latency in the MWM task in the STZ + ICSS group compared to untreated STZ rats at 5 months post-STZ. At Ser0, levels of miR-16, miR-30c, miR-181, miR-191 and miR-196a were significantly increased in STZ group. In STZ rats, miR-146a and miR-495 levels increased from Ser1 to Ser2, an effect not observed in the Control or STZ + ICSS groups. In SH-SY5Y cells, miR-495 overexpression significantly downregulated both NRF2 mRNA and protein levels. Moreover, STZ exposure increased miR-495 and reduced NRF2 protein levels. MFB-ICSS also reversed the STZ-induced reductions in both CA1 NRF2 levels and corpus callosum thickness.
CONCLUSION: Prolonged MFB-ICSS treatment mitigates cognitive deficits, modulates circulating levels of miRNA-495 and miR-146a, restores hippocampal NRF2 levels, and preserves corpus callosum integrity in the SAD rat model by STZ injection. These findings highlight the therapeutic potential of MFB-ICSS as a non-pharmacological intervention in AD. Furthermore, this study confirms NRF2 as a target of miR-495 in the context of AD.
Additional Links: PMID-41306402
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306402,
year = {2025},
author = {Riberas-Sánchez, A and García-Brito, S and Vila-Solés, L and Aldavert-Vera, L and Segura-Torres, P and Huguet, G and Carreras-Badosa, G and Kádár, E},
title = {Intracranial self-stimulation mitigates spatial task deficits, modifies miR-146a and miR-495 serum levels and restores hippocampal NRF2 levels in a rat model of sporadic Alzheimer's disease.},
journal = {Frontiers in aging neuroscience},
volume = {17},
number = {},
pages = {1671196},
pmid = {41306402},
issn = {1663-4365},
abstract = {INTRODUCTION: Alzheimer's disease (AD) is the most prevalent neurodegenerative disease associated with aging. While deep brain stimulation (DBS) shows therapeutic promise, the long-term persistence of its effects remains understudied. Expression patterns of circulating miRNAs, proposed diagnostic biomarkers, and their modulation by DBS are still poorly characterized in longitudinal studies. This study investigates the effect of a 13-week prolonged ICSS treatment on spatial memory and serum miRNA expression in a male rat model of sporadic AD (SAD) by intracerebroventricular injection of streptozotocin (STZ).
METHODS: Morris water maze (MWM) tasks were conducted at 1 and 5 months post-STZ. Serum miRNA levels were quantified by qRT-PCR at 29 (Ser0), 73 (Ser1) and 136 (Ser2) days after STZ administration. Corpus callosum thickness and NRF2 protein levels in the hippocampal CA1 region were assessed using Nissl staining and immunohistochemistry, respectively. Target validation of miR-495 was performed via transfection assays in the human neuroblastoma SH-SY5Y cell line.
RESULTS: MFB-ICSS treatment significantly reduced escape latency in the MWM task in the STZ + ICSS group compared to untreated STZ rats at 5 months post-STZ. At Ser0, levels of miR-16, miR-30c, miR-181, miR-191 and miR-196a were significantly increased in STZ group. In STZ rats, miR-146a and miR-495 levels increased from Ser1 to Ser2, an effect not observed in the Control or STZ + ICSS groups. In SH-SY5Y cells, miR-495 overexpression significantly downregulated both NRF2 mRNA and protein levels. Moreover, STZ exposure increased miR-495 and reduced NRF2 protein levels. MFB-ICSS also reversed the STZ-induced reductions in both CA1 NRF2 levels and corpus callosum thickness.
CONCLUSION: Prolonged MFB-ICSS treatment mitigates cognitive deficits, modulates circulating levels of miRNA-495 and miR-146a, restores hippocampal NRF2 levels, and preserves corpus callosum integrity in the SAD rat model by STZ injection. These findings highlight the therapeutic potential of MFB-ICSS as a non-pharmacological intervention in AD. Furthermore, this study confirms NRF2 as a target of miR-495 in the context of AD.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Clarifying blood-based brain-derived neurotrophic factor measurement in Alzheimer's disease research: A call for pre-analytic standardization.
Journal of Alzheimer's disease reports, 9:25424823251399060.
The study by Kueck et al. rigorously addresses the complex pre-analytical and biological confounds underlying blood-based brain-derived neurotrophic factor (BDNF) measurement in Alzheimer's disease (AD) research. By directly comparing platelet-rich plasma and platelet-poor plasma, it distinguishes free-circulating from platelet-stored BDNF and their associations with core AD biomarkers. Findings support BDNF as a potential compensatory marker in early cognitive impairment and emphasize the critical need for biomarker-specific pre-analytic standardization protocols. This commentary contextualizes the work within existing inconsistencies and outlines key recommendations for standardizing blood-based BDNF measurements to ensure reproducibility, potential clinical utility, and translational relevance.
Additional Links: PMID-41306381
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306381,
year = {2025},
author = {Ng, TK},
title = {Clarifying blood-based brain-derived neurotrophic factor measurement in Alzheimer's disease research: A call for pre-analytic standardization.},
journal = {Journal of Alzheimer's disease reports},
volume = {9},
number = {},
pages = {25424823251399060},
pmid = {41306381},
issn = {2542-4823},
abstract = {The study by Kueck et al. rigorously addresses the complex pre-analytical and biological confounds underlying blood-based brain-derived neurotrophic factor (BDNF) measurement in Alzheimer's disease (AD) research. By directly comparing platelet-rich plasma and platelet-poor plasma, it distinguishes free-circulating from platelet-stored BDNF and their associations with core AD biomarkers. Findings support BDNF as a potential compensatory marker in early cognitive impairment and emphasize the critical need for biomarker-specific pre-analytic standardization protocols. This commentary contextualizes the work within existing inconsistencies and outlines key recommendations for standardizing blood-based BDNF measurements to ensure reproducibility, potential clinical utility, and translational relevance.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Invisible Invaders: How Nanoplastics Hijack the Brain and Accelerate Alzheimer's Pathology.
Environment & health (Washington, D.C.), 3(11):1264-1265.
Additional Links: PMID-41306308
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306308,
year = {2025},
author = {Sun, H and Song, Y},
title = {Invisible Invaders: How Nanoplastics Hijack the Brain and Accelerate Alzheimer's Pathology.},
journal = {Environment & health (Washington, D.C.)},
volume = {3},
number = {11},
pages = {1264-1265},
pmid = {41306308},
issn = {2833-8278},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Cerebral to Systemic Representations of Alzheimer's Pathogenesis Stimulated by Polystyrene Nanoplastics.
Environment & health (Washington, D.C.), 3(11):1380-1392.
Plastics discharged into the ecosphere can transform into micro- and nanoparticles to instigate interactions with biosystems, posing a threat to environmental sustainability and human health. While nanoplastics have recently been identified in abundance in the human brain, especially in the decedent brain tissues of dementia subjects, how these exogenous miniatures mediate neurological as well as systemic pathologies remains unclear. Here, we first investigated how environmental-level nanoplastic exposure influences the progression of Alzheimer's disease, from cerebral to systemic representations. Specifically, polystyrene nanoplastics aggravated Alzheimer's-like symptoms in both wild-type and APP/PS1 mice and stimulated microglial activation and hippocampal neuronal death, accentuated by peripheral abnormalities of lipid accumulation, hepatic steatosis, inflammation, adipocyte enlargement, and gut microbiota imbalance. These findings implicate that nanoplastic-induced neurological damage is not confined within the brain but expands systemically through the gut-liver-brain axis, thereby contributing to the multiscale and multidirectional progression of Alzheimer's pathophysiology.
Additional Links: PMID-41306300
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306300,
year = {2025},
author = {Wang, Y and Liang, X and Andrikopoulos, N and Zheng, S and Li, Y and Ke, PC},
title = {Cerebral to Systemic Representations of Alzheimer's Pathogenesis Stimulated by Polystyrene Nanoplastics.},
journal = {Environment & health (Washington, D.C.)},
volume = {3},
number = {11},
pages = {1380-1392},
pmid = {41306300},
issn = {2833-8278},
abstract = {Plastics discharged into the ecosphere can transform into micro- and nanoparticles to instigate interactions with biosystems, posing a threat to environmental sustainability and human health. While nanoplastics have recently been identified in abundance in the human brain, especially in the decedent brain tissues of dementia subjects, how these exogenous miniatures mediate neurological as well as systemic pathologies remains unclear. Here, we first investigated how environmental-level nanoplastic exposure influences the progression of Alzheimer's disease, from cerebral to systemic representations. Specifically, polystyrene nanoplastics aggravated Alzheimer's-like symptoms in both wild-type and APP/PS1 mice and stimulated microglial activation and hippocampal neuronal death, accentuated by peripheral abnormalities of lipid accumulation, hepatic steatosis, inflammation, adipocyte enlargement, and gut microbiota imbalance. These findings implicate that nanoplastic-induced neurological damage is not confined within the brain but expands systemically through the gut-liver-brain axis, thereby contributing to the multiscale and multidirectional progression of Alzheimer's pathophysiology.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Control analysis of deep brain stimulation and optogenetics for Alzheimer's disease under the computational cortex model.
Cognitive neurodynamics, 20(1):10.
Abnormal τ and β-amyloid (Aβ) deposition in the brains of patients with Alzheimer's disease (AD) is significantly associated with cognitive decline. This abnormal deposition has been reported to be linked to increased excitatory and inhibitory time constants in neural circuits. In this paper, we focus on three typical electroencephalography (EEG) slowdowns clinically reported in association with AD, including decreased dominant frequency, decreased α rhythmic activity, and increased δ + θ rhythmic activity. Firstly, we demonstrate that changes in excitatory time constant, inhibitory time constants, and synaptic connection strength can induce the phenomenon of EEG slowdowns in early AD. Then, we are interested in the regulation of AD by traditional deep brain stimulation (DBS) and emerging optogenetic stimulation. High-frequency, high-pulse width, and high-amplitude DBS are more effective in reversing brain rhythm in AD, supporting the experiment that cortical high-frequency DBS may be an effective therapeutic way for dementia-related diseases. In particular, as a modification of traditional DBS, we find that oscillatory bursty stimulation can compensate for the shortcomings of DBS at low amplitude. However, it is physiologically difficult to target inhibitory interneurons with conventional electrical stimulation. Optogenetics is able to precisely stimulate pyramidal neurons and inhibitory interneurons observed in animal experiments. Our numerical results indicate that medium and low-frequency stimulation can better eliminate AD pathology. It should be noted that stimulation of inhibitory interneurons requires greater light intensity than stimulation of pyramidal neurons. Finally, we propose two optimization intermittent optogenetic stimulation protocols. These modeling results can reproduce some experimental phenomena and are expected to reveal the underlying pathological mechanisms and control strategies associated with cognitive dysfunction such as AD.
Additional Links: PMID-41306192
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306192,
year = {2026},
author = {Zhang, Y and Zhang, H and Shen, Z},
title = {Control analysis of deep brain stimulation and optogenetics for Alzheimer's disease under the computational cortex model.},
journal = {Cognitive neurodynamics},
volume = {20},
number = {1},
pages = {10},
pmid = {41306192},
issn = {1871-4080},
abstract = {Abnormal τ and β-amyloid (Aβ) deposition in the brains of patients with Alzheimer's disease (AD) is significantly associated with cognitive decline. This abnormal deposition has been reported to be linked to increased excitatory and inhibitory time constants in neural circuits. In this paper, we focus on three typical electroencephalography (EEG) slowdowns clinically reported in association with AD, including decreased dominant frequency, decreased α rhythmic activity, and increased δ + θ rhythmic activity. Firstly, we demonstrate that changes in excitatory time constant, inhibitory time constants, and synaptic connection strength can induce the phenomenon of EEG slowdowns in early AD. Then, we are interested in the regulation of AD by traditional deep brain stimulation (DBS) and emerging optogenetic stimulation. High-frequency, high-pulse width, and high-amplitude DBS are more effective in reversing brain rhythm in AD, supporting the experiment that cortical high-frequency DBS may be an effective therapeutic way for dementia-related diseases. In particular, as a modification of traditional DBS, we find that oscillatory bursty stimulation can compensate for the shortcomings of DBS at low amplitude. However, it is physiologically difficult to target inhibitory interneurons with conventional electrical stimulation. Optogenetics is able to precisely stimulate pyramidal neurons and inhibitory interneurons observed in animal experiments. Our numerical results indicate that medium and low-frequency stimulation can better eliminate AD pathology. It should be noted that stimulation of inhibitory interneurons requires greater light intensity than stimulation of pyramidal neurons. Finally, we propose two optimization intermittent optogenetic stimulation protocols. These modeling results can reproduce some experimental phenomena and are expected to reveal the underlying pathological mechanisms and control strategies associated with cognitive dysfunction such as AD.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Risk prediction of mild cognitive impairment using electronic health record data.
Alzheimer's & dementia (Amsterdam, Netherlands), 17(4):e70209.
INTRODUCTION: Mild cognitive impairment (MCI) is underdiagnosed by primary care providers (PCPs), with detection rates as low as 6%-15%. Predictive models support the identification of individuals at risk, enabling timely intervention.
METHODS: This retrospective study was conducted on 271,054 cognitively unimpaired and 14,501 confirmed MCI cohorts from electronic health records. A machine learning model was developed with a data-driven variable selection approach based on demographics and comorbidities.
RESULTS: From 101 variables, 26 were chosen for the final model, achieving an overall area under the curve (AUC) of 86%. Age-stratified AUCs were 79.1% (40-49), 77.0% (50-64), 76.9% (65-79), and 74.4% (≥80), showing the highest predictive performance in younger age groups.
DISCUSSION: Demographic factors and comorbidities can serve as effective predictors for the risk of MCI. The model demonstrates strong predictive performance and assists as a triage tool for PCPs, facilitating the identification of individuals with MCI for early treatment.
HIGHLIGHTS: Predictive algorithms using electronic health records (EHRs) are useful for identifying individuals at risk for mild cognitive impairment (MCI) to triage for further clinical evaluation.A machine learning model was developed using EHR data to predict those at risk for MCI.The model identified 26 variables that were able to distinguish the MCI from non-MCI cohorts.The model accurately detected MCI across the cohort (area under the curve [AUC] = 86%) and trended best for younger age groups (AUC was 77%, 77%, and 74% in 50-64, 65-79, and ≥80 age groups, respectively).Implementation of a triage tool could be used to detect MCI across aging patient populations sooner, leading to a timelier diagnosis, intervention, and treatment management.
Additional Links: PMID-41306115
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41306115,
year = {2025},
author = {Li, G and Cobb, B and Nelson, TM and Devanarayan, V and Borentain, S and Mielke, MM and Galvin, JE and Kivipelto, M and Tkatch, R and De Santi, S and Frech, F and Vandercappellen, J and Nakamura, Y and Crislip, R and Meyerhoff, J and Mattke, S and Hampel, H},
title = {Risk prediction of mild cognitive impairment using electronic health record data.},
journal = {Alzheimer's & dementia (Amsterdam, Netherlands)},
volume = {17},
number = {4},
pages = {e70209},
pmid = {41306115},
issn = {2352-8729},
abstract = {INTRODUCTION: Mild cognitive impairment (MCI) is underdiagnosed by primary care providers (PCPs), with detection rates as low as 6%-15%. Predictive models support the identification of individuals at risk, enabling timely intervention.
METHODS: This retrospective study was conducted on 271,054 cognitively unimpaired and 14,501 confirmed MCI cohorts from electronic health records. A machine learning model was developed with a data-driven variable selection approach based on demographics and comorbidities.
RESULTS: From 101 variables, 26 were chosen for the final model, achieving an overall area under the curve (AUC) of 86%. Age-stratified AUCs were 79.1% (40-49), 77.0% (50-64), 76.9% (65-79), and 74.4% (≥80), showing the highest predictive performance in younger age groups.
DISCUSSION: Demographic factors and comorbidities can serve as effective predictors for the risk of MCI. The model demonstrates strong predictive performance and assists as a triage tool for PCPs, facilitating the identification of individuals with MCI for early treatment.
HIGHLIGHTS: Predictive algorithms using electronic health records (EHRs) are useful for identifying individuals at risk for mild cognitive impairment (MCI) to triage for further clinical evaluation.A machine learning model was developed using EHR data to predict those at risk for MCI.The model identified 26 variables that were able to distinguish the MCI from non-MCI cohorts.The model accurately detected MCI across the cohort (area under the curve [AUC] = 86%) and trended best for younger age groups (AUC was 77%, 77%, and 74% in 50-64, 65-79, and ≥80 age groups, respectively).Implementation of a triage tool could be used to detect MCI across aging patient populations sooner, leading to a timelier diagnosis, intervention, and treatment management.},
}
RevDate: 2025-11-27
In-Situ Spatial Visual Proteomics Enabled by Single-Cell-Type Proximity Biotinylation and Signaling Amplification.
Angewandte Chemie (International ed. in English) [Epub ahead of print].
Mapping the spatial proteome signature within heterogeneous tissue microenvironment of clinical specimens offers unique insights into both physiological and pathological molecular phenotypes. However, to simultaneously visualize and discover these spatial proteome features is challenging. Herein, we introduce spatial visual proteomics (SVPro) which integrates single-cell proximity biotinylation coupled with fluorescence signal amplification via the trifunctional probe. This probe uniquely combines naphthylamine-mediated radical targeting, clickable fluorescence imaging, and biotinylation-facilitated proteome capture, enabling all three within a single tissue section. Strategic PEGylation and multivalent fluorescent conjugation of these probes improves labeling and visualization efficiency and specificity. SVPro allows cell-type proteomic profiling of neuronal subpopulations in distinct mouse brain regions and amyloid-β plaques in mouse brain of Alzheimer's disease model. Notably, we identified and validated distinct marker proteins associated with amyloid‑β plaques deposited in different brain regions. SVPro therefore emerges as a robust and high-resolution spatial proteomics approach for simultaneously visualizing the spatial architecture and proteome landscapes of intact tissue microenvironment.
Additional Links: PMID-41305902
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41305902,
year = {2025},
author = {Zheng, Z and Tang, Z and Li, C and Xiao, H and Li, Y and He, A and Mao, Y and Zheng, J and Ke, M and Gao, R and Li, D and Liu, C and Dong, Z and Tian, R},
title = {In-Situ Spatial Visual Proteomics Enabled by Single-Cell-Type Proximity Biotinylation and Signaling Amplification.},
journal = {Angewandte Chemie (International ed. in English)},
volume = {},
number = {},
pages = {e19440},
doi = {10.1002/anie.202519440},
pmid = {41305902},
issn = {1521-3773},
abstract = {Mapping the spatial proteome signature within heterogeneous tissue microenvironment of clinical specimens offers unique insights into both physiological and pathological molecular phenotypes. However, to simultaneously visualize and discover these spatial proteome features is challenging. Herein, we introduce spatial visual proteomics (SVPro) which integrates single-cell proximity biotinylation coupled with fluorescence signal amplification via the trifunctional probe. This probe uniquely combines naphthylamine-mediated radical targeting, clickable fluorescence imaging, and biotinylation-facilitated proteome capture, enabling all three within a single tissue section. Strategic PEGylation and multivalent fluorescent conjugation of these probes improves labeling and visualization efficiency and specificity. SVPro allows cell-type proteomic profiling of neuronal subpopulations in distinct mouse brain regions and amyloid-β plaques in mouse brain of Alzheimer's disease model. Notably, we identified and validated distinct marker proteins associated with amyloid‑β plaques deposited in different brain regions. SVPro therefore emerges as a robust and high-resolution spatial proteomics approach for simultaneously visualizing the spatial architecture and proteome landscapes of intact tissue microenvironment.},
}
RevDate: 2025-11-27
Hydroxycarboxylic Acid Receptor 2 Mediates β-hydroxybutyrate's Antiseizure Effect in Mice.
Annals of neurology [Epub ahead of print].
OBJECTIVE: The ketogenic diet, a high-fat, low-carbohydrate regimen, is often used to treat drug-resistant seizures and is being studied for Alzheimer's disease and other neuropsychiatric disorders. However, its mechanism of action remains unclear. β-hydroxybutyrate, a primary circulating ketone body produced by the ketogenic diet, may mediate its effects on seizures by binding to a recently identified Gi-coupled receptor: hydrocarboxylic acid receptor 2 (HCAR2).
METHODS: RNAscope in situ hybridization assay and real-time quantitative polymerase chain reaction were used to assess HCAR2 expression in the mouse brain. We generated HCAR2[-]/[-] using the CRISPR-Cas technique on an S129 mouse background. Whole-cell current-clamp was performed to measure the passive and active membrane properties of hippocampal dentate granule cells. The voltage-clamp was performed to record synaptic currents. Two complementary in vivo mouse models-continuous hippocampal stimulation to induce status epilepticus (SE) and kindling-were used to induce seizures.
RESULTS: HCAR2 was localized in dentate granule cells and microglia. In mice with HCAR2, β-hydroxybutyrate reduced neuronal excitability by hyperpolarizing the resting membrane potential, raising the action potential threshold, and reducing the firing frequency of dentate granule cells. β-hydroxybutyrate suppressed excitatory synaptic transmission. These effects were nullified in HCAR2[-]/[-] mice. HCAR2[-]/[-] mice showed no cognitive impairment. Moreover, β-hydroxybutyrate did not affect seizures in HCAR2[-]/[-] mice. However, it diminished both the duration and severity of seizures in HCAR2[+]/[+] mice.
INTERPRETATION: These findings demonstrate that HCAR2 mediates β-hydroxybutyrate's antiseizure effects by regulating neuronal excitability and synaptic transmission. These studies propose a new mechanism for the antiseizure action of the ketogenic diet. ANN NEUROL 2025.
Additional Links: PMID-41305866
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41305866,
year = {2025},
author = {Naderi, S and Williamson, J and Sun, H and Joshi, S and Spera, RJ and Zaib, S and Sharma, S and Sun, C and Brodovskiy, A and Zawar, I and Kapur, J},
title = {Hydroxycarboxylic Acid Receptor 2 Mediates β-hydroxybutyrate's Antiseizure Effect in Mice.},
journal = {Annals of neurology},
volume = {},
number = {},
pages = {},
doi = {10.1002/ana.78098},
pmid = {41305866},
issn = {1531-8249},
support = {R01NS120945//United States National Institute of Health (NINDS)/ ; R37N119012//United States National Institute of Health (NINDS)/ ; },
abstract = {OBJECTIVE: The ketogenic diet, a high-fat, low-carbohydrate regimen, is often used to treat drug-resistant seizures and is being studied for Alzheimer's disease and other neuropsychiatric disorders. However, its mechanism of action remains unclear. β-hydroxybutyrate, a primary circulating ketone body produced by the ketogenic diet, may mediate its effects on seizures by binding to a recently identified Gi-coupled receptor: hydrocarboxylic acid receptor 2 (HCAR2).
METHODS: RNAscope in situ hybridization assay and real-time quantitative polymerase chain reaction were used to assess HCAR2 expression in the mouse brain. We generated HCAR2[-]/[-] using the CRISPR-Cas technique on an S129 mouse background. Whole-cell current-clamp was performed to measure the passive and active membrane properties of hippocampal dentate granule cells. The voltage-clamp was performed to record synaptic currents. Two complementary in vivo mouse models-continuous hippocampal stimulation to induce status epilepticus (SE) and kindling-were used to induce seizures.
RESULTS: HCAR2 was localized in dentate granule cells and microglia. In mice with HCAR2, β-hydroxybutyrate reduced neuronal excitability by hyperpolarizing the resting membrane potential, raising the action potential threshold, and reducing the firing frequency of dentate granule cells. β-hydroxybutyrate suppressed excitatory synaptic transmission. These effects were nullified in HCAR2[-]/[-] mice. HCAR2[-]/[-] mice showed no cognitive impairment. Moreover, β-hydroxybutyrate did not affect seizures in HCAR2[-]/[-] mice. However, it diminished both the duration and severity of seizures in HCAR2[+]/[+] mice.
INTERPRETATION: These findings demonstrate that HCAR2 mediates β-hydroxybutyrate's antiseizure effects by regulating neuronal excitability and synaptic transmission. These studies propose a new mechanism for the antiseizure action of the ketogenic diet. ANN NEUROL 2025.},
}
RevDate: 2025-11-27
CmpDate: 2025-11-27
Alzheimer's and Parkinson's diseases in relation to human papillomavirus infection: A two-sample Mendelian randomization study.
Medicine, 104(47):e46043.
An increasing number of studies are exploring links between human papillomavirus (HPV) infection and neurodegenerative diseases. Yet, existing evidence from retrospective studies is prone to bias and cannot confirm causation. To address this, our 2-sample Mendelian randomization (MR) study utilizing single nucleoside polymers aims to establish a potential causal connection between HPV and the onset of Alzheimer disease (AD) and Parkinson disease (PD). We utilized a 2-sample MR approach to evaluate the causal relationships between HPV16 and HPV18 E7 proteins, HPV seropositivity, and the risks of AD and PD. Selected single nucleoside polymers from genome-wide association studies served as instrumental variables in HPV exposure data for MR analysis. Subsequent rigorous sensitivity assessments were also performed to ensure robustness. In this research, we explored the potential causal connections between HPV infection and the development of AD or PD through a MR analysis. Our findings suggest that the HPV18 E7 protein might act as a protective factor against PD, whereas HPV seropositivity was recognized as a risk factor for the same condition. Interestingly, neither the HPV16 E7 protein nor HPV seropositivity appeared to have any significant impact on the risk of AD. These findings shed light on HPV's intricate involvement in neurodegenerative diseases, offering fresh views on viral impacts on disorders like PD and AD. Recognizing study limitations, further research is crucial to confirm and clarify HPV's exact effects on these conditions.
Additional Links: PMID-41305738
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41305738,
year = {2025},
author = {Chen, B and Chen, Y and Zhang, J and Yang, W and Xu, Y and Chen, X and Chen, N and Xia, J and Xia, J and Ding, H and Yang, T},
title = {Alzheimer's and Parkinson's diseases in relation to human papillomavirus infection: A two-sample Mendelian randomization study.},
journal = {Medicine},
volume = {104},
number = {47},
pages = {e46043},
doi = {10.1097/MD.0000000000046043},
pmid = {41305738},
issn = {1536-5964},
mesh = {Humans ; Mendelian Randomization Analysis ; *Alzheimer Disease/virology/genetics/epidemiology ; *Parkinson Disease/virology/genetics/epidemiology ; *Papillomavirus Infections/complications/epidemiology/genetics ; Genome-Wide Association Study ; Risk Factors ; Papillomavirus E7 Proteins/blood ; Female ; Male ; Human papillomavirus 16 ; Human papillomavirus 18 ; },
abstract = {An increasing number of studies are exploring links between human papillomavirus (HPV) infection and neurodegenerative diseases. Yet, existing evidence from retrospective studies is prone to bias and cannot confirm causation. To address this, our 2-sample Mendelian randomization (MR) study utilizing single nucleoside polymers aims to establish a potential causal connection between HPV and the onset of Alzheimer disease (AD) and Parkinson disease (PD). We utilized a 2-sample MR approach to evaluate the causal relationships between HPV16 and HPV18 E7 proteins, HPV seropositivity, and the risks of AD and PD. Selected single nucleoside polymers from genome-wide association studies served as instrumental variables in HPV exposure data for MR analysis. Subsequent rigorous sensitivity assessments were also performed to ensure robustness. In this research, we explored the potential causal connections between HPV infection and the development of AD or PD through a MR analysis. Our findings suggest that the HPV18 E7 protein might act as a protective factor against PD, whereas HPV seropositivity was recognized as a risk factor for the same condition. Interestingly, neither the HPV16 E7 protein nor HPV seropositivity appeared to have any significant impact on the risk of AD. These findings shed light on HPV's intricate involvement in neurodegenerative diseases, offering fresh views on viral impacts on disorders like PD and AD. Recognizing study limitations, further research is crucial to confirm and clarify HPV's exact effects on these conditions.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Mendelian Randomization Analysis
*Alzheimer Disease/virology/genetics/epidemiology
*Parkinson Disease/virology/genetics/epidemiology
*Papillomavirus Infections/complications/epidemiology/genetics
Genome-Wide Association Study
Risk Factors
Papillomavirus E7 Proteins/blood
Female
Male
Human papillomavirus 16
Human papillomavirus 18
RevDate: 2025-11-27
CmpDate: 2025-11-27
Future Directions in Choline: From Neurodevelopment to Cardiometabolic Health.
Nutrients, 17(22): pii:nu17223618.
Although choline was established as an essential nutrient over three decades ago, critical questions remain about how choline regulates brain, liver, and cardiometabolic health across the lifespan. This Perspective summarizes emerging insights presented at the Future Directions in Choline Symposium: A Tribute to Steven H. Zeisel, which outlined three converging areas of research: (i) choline-dependent mechanisms in neurodevelopment and cognition, (ii) the link between choline metabolism and obesity, and (iii) the role of trimethylamine N-oxide (TMAO) in cardiovascular disease. Evidence from clinical and preclinical studies confirms that maternal choline intake is critical for neurogenesis, cognition, and visual system development, and that higher choline availability buffers the fetal brain against environmental and psychosocial stressors. Choline supplementation improves cognitive outcomes in fetal alcohol spectrum disorder and mitigates neurodegenerative pathology in Alzheimer's models. In cardiometabolic health, recent data challenge the interpretation of TMAO as a causal toxin, positioning it instead as a marker of renal function. Moving forward, the field must develop validated biomarkers of choline adequacy in free-living populations, harmonize intervention protocols, and define context-specific requirements across obesity, pregnancy, and glucagon-like peptide-1 (GLP-1)-based therapy use. These efforts will refine dietary recommendations and solidify choline's role in lifelong brain and metabolic health.
Additional Links: PMID-41305668
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41305668,
year = {2025},
author = {Paules, EM and Petry, HG and Sprinkles, JK and Trujillo-Gonzalez, I},
title = {Future Directions in Choline: From Neurodevelopment to Cardiometabolic Health.},
journal = {Nutrients},
volume = {17},
number = {22},
pages = {},
doi = {10.3390/nu17223618},
pmid = {41305668},
issn = {2072-6643},
support = {N/A//Nutrition Research Institute, start up to I. T-G/ ; R01 DK115380/DK/NIDDK NIH HHS/United States ; },
mesh = {Humans ; *Choline/metabolism/administration & dosage ; Pregnancy ; *Cardiovascular Diseases/prevention & control/metabolism ; Female ; Brain/metabolism/drug effects ; Cognition/drug effects ; Obesity/metabolism ; Methylamines/metabolism ; Animals ; Dietary Supplements ; },
abstract = {Although choline was established as an essential nutrient over three decades ago, critical questions remain about how choline regulates brain, liver, and cardiometabolic health across the lifespan. This Perspective summarizes emerging insights presented at the Future Directions in Choline Symposium: A Tribute to Steven H. Zeisel, which outlined three converging areas of research: (i) choline-dependent mechanisms in neurodevelopment and cognition, (ii) the link between choline metabolism and obesity, and (iii) the role of trimethylamine N-oxide (TMAO) in cardiovascular disease. Evidence from clinical and preclinical studies confirms that maternal choline intake is critical for neurogenesis, cognition, and visual system development, and that higher choline availability buffers the fetal brain against environmental and psychosocial stressors. Choline supplementation improves cognitive outcomes in fetal alcohol spectrum disorder and mitigates neurodegenerative pathology in Alzheimer's models. In cardiometabolic health, recent data challenge the interpretation of TMAO as a causal toxin, positioning it instead as a marker of renal function. Moving forward, the field must develop validated biomarkers of choline adequacy in free-living populations, harmonize intervention protocols, and define context-specific requirements across obesity, pregnancy, and glucagon-like peptide-1 (GLP-1)-based therapy use. These efforts will refine dietary recommendations and solidify choline's role in lifelong brain and metabolic health.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Choline/metabolism/administration & dosage
Pregnancy
*Cardiovascular Diseases/prevention & control/metabolism
Female
Brain/metabolism/drug effects
Cognition/drug effects
Obesity/metabolism
Methylamines/metabolism
Animals
Dietary Supplements
RevDate: 2025-11-27
CmpDate: 2025-11-27
The Role of Bacopa monnieri in Alzheimer's Disease: Mechanisms and Potential Clinical Use-A Review.
Nutrients, 17(22): pii:nu17223538.
Over the past few years, there has been a growing interest in traditional Ayurvedic medicine, which extensively utilizes plants. One such plant is Bacopa monnieri, also known as brahmi, which has gained particular popularity for its cognitive-function-enhancing properties and neuroprotective effects. Numerous studies highlight its antioxidant, anti-inflammatory, anti-stress, anti-anxiety, and even symptom-reducing properties associated with attention deficit-hyperactivity disorder (ADHD). Additionally, in vitro and in vivo studies demonstrate the potential therapeutic applications of this plant and its active compounds, particularly bacosides, in treating Alzheimer's disease. This review aims to evaluate whether B. monnieri can serve as a potential therapy enhancer and slow the progression of Alzheimer's disease. We have attempted to clarify the mechanisms of action and the possible clinical application of brahmi in this progressive neurodegenerative brain disorder.
Additional Links: PMID-41305589
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid41305589,
year = {2025},
author = {Wiciński, M and Fajkiel-Madajczyk, A and Wójcicki, J and Ozorowski, M and Szambelan, M},
title = {The Role of Bacopa monnieri in Alzheimer's Disease: Mechanisms and Potential Clinical Use-A Review.},
journal = {Nutrients},
volume = {17},
number = {22},
pages = {},
doi = {10.3390/nu17223538},
pmid = {41305589},
issn = {2072-6643},
mesh = {*Bacopa/chemistry ; *Alzheimer Disease/drug therapy ; Humans ; *Neuroprotective Agents/pharmacology/therapeutic use ; *Plant Extracts/pharmacology/therapeutic use ; Animals ; Medicine, Ayurvedic ; Phytotherapy ; Antioxidants/pharmacology ; },
abstract = {Over the past few years, there has been a growing interest in traditional Ayurvedic medicine, which extensively utilizes plants. One such plant is Bacopa monnieri, also known as brahmi, which has gained particular popularity for its cognitive-function-enhancing properties and neuroprotective effects. Numerous studies highlight its antioxidant, anti-inflammatory, anti-stress, anti-anxiety, and even symptom-reducing properties associated with attention deficit-hyperactivity disorder (ADHD). Additionally, in vitro and in vivo studies demonstrate the potential therapeutic applications of this plant and its active compounds, particularly bacosides, in treating Alzheimer's disease. This review aims to evaluate whether B. monnieri can serve as a potential therapy enhancer and slow the progression of Alzheimer's disease. We have attempted to clarify the mechanisms of action and the possible clinical application of brahmi in this progressive neurodegenerative brain disorder.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Bacopa/chemistry
*Alzheimer Disease/drug therapy
Humans
*Neuroprotective Agents/pharmacology/therapeutic use
*Plant Extracts/pharmacology/therapeutic use
Animals
Medicine, Ayurvedic
Phytotherapy
Antioxidants/pharmacology
▼ ▼ LOAD NEXT 100 CITATIONS
RJR Experience and Expertise
Researcher
Robbins holds BS, MS, and PhD degrees in the life sciences. He served as a tenured faculty member in the Zoology and Biological Science departments at Michigan State University. He is currently exploring the intersection between genomics, microbial ecology, and biodiversity — an area that promises to transform our understanding of the biosphere.
Educator
Robbins has extensive experience in college-level education: At MSU he taught introductory biology, genetics, and population genetics. At JHU, he was an instructor for a special course on biological database design. At FHCRC, he team-taught a graduate-level course on the history of genetics. At Bellevue College he taught medical informatics.
Administrator
Robbins has been involved in science administration at both the federal and the institutional levels. At NSF he was a program officer for database activities in the life sciences, at DOE he was a program officer for information infrastructure in the human genome project. At the Fred Hutchinson Cancer Research Center, he served as a vice president for fifteen years.
Technologist
Robbins has been involved with information technology since writing his first Fortran program as a college student. At NSF he was the first program officer for database activities in the life sciences. At JHU he held an appointment in the CS department and served as director of the informatics core for the Genome Data Base. At the FHCRC he was VP for Information Technology.
Publisher
While still at Michigan State, Robbins started his first publishing venture, founding a small company that addressed the short-run publishing needs of instructors in very large undergraduate classes. For more than 20 years, Robbins has been operating The Electronic Scholarly Publishing Project, a web site dedicated to the digital publishing of critical works in science, especially classical genetics.
Speaker
Robbins is well-known for his speaking abilities and is often called upon to provide keynote or plenary addresses at international meetings. For example, in July, 2012, he gave a well-received keynote address at the Global Biodiversity Informatics Congress, sponsored by GBIF and held in Copenhagen. The slides from that talk can be seen HERE.
Facilitator
Robbins is a skilled meeting facilitator. He prefers a participatory approach, with part of the meeting involving dynamic breakout groups, created by the participants in real time: (1) individuals propose breakout groups; (2) everyone signs up for one (or more) groups; (3) the groups with the most interested parties then meet, with reports from each group presented and discussed in a subsequent plenary session.
Designer
Robbins has been engaged with photography and design since the 1960s, when he worked for a professional photography laboratory. He now prefers digital photography and tools for their precision and reproducibility. He designed his first web site more than 20 years ago and he personally designed and implemented this web site. He engages in graphic design as a hobby.
RJR Picks from Around the Web (updated 11 MAY 2018 )
Old Science
Weird Science
Treating Disease with Fecal Transplantation
Fossils of miniature humans (hobbits) discovered in Indonesia
Paleontology
Dinosaur tail, complete with feathers, found preserved in amber.
Astronomy
Mysterious fast radio burst (FRB) detected in the distant universe.
Big Data & Informatics
Big Data: Buzzword or Big Deal?
Hacking the genome: Identifying anonymized human subjects using publicly available data.