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 24 Apr 2025 at 01:37 Created:
Alzheimer Disease — Treatment
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. Because of this lack of understanding of the root cause for Alzheimer's Disease, no direct treatment for the condition is yet available. However, this bibliography specifically searches for the idea of treatment in conjunction with Alzheimer's to make it easier to track literature that explores the possibility of treatment.
Created with PubMed® Query: ( alzheimer*[TIAB] AND treatment[TIAB] ) NOT pmcbook NOT ispreviousversion
Citations The Papers (from PubMed®)
RevDate: 2025-04-23
Blockage of CCL3 with neutralizing antibody reduces neuroinflammation and reverses Alzheimer disease phenotypes.
Brain, behavior, and immunity pii:S0889-1591(25)00171-0 [Epub ahead of print].
BACKGROUND: Accumulating evidence indicates that neuroinflammation is involved in the pathogenesis of Alzheimer's disease (AD). According to RNA sequencing and quantitative PCR (qPCR), we found that chemokine CCL3 mRNA expression was abnormally upregulated in the brains of AD transgenic mice. Moreover, the levels of CCL3 in the serum of AD patients were significantly elevated and negatively correlated with their cognitive abilities. However, the role of CCL3 in AD neuroinflammation and pathological damages remains elusive.
METHODS: Using behavioral, histological, and biochemical methods, outcomes of CCL3 antibody treatment on neuropathology and cognitive deficits were studied in the APPswe/PS1dE9 mice.
RESULTS: In the present study, we reported that CCL3 protein expression was increased in the APPswe/PS1dE9 mice, whereas blockage of CCL3 with neutralizing antibody potently inhibited CCL3 activation in the APPswe/PS1dE9 mice down to the levels of wild-type mice. Specifically, CCL3 antibody significantly improved the learning and memory abilities of APPswe/PS1dE9 mice. In addition, CCL3 antibody treatment decreased cerebral amyloid-β (Aβ) levels and plaque burden via inhibiting amyloid precursor protein (APP) processing by reducing beta-site APP cleaving enzyme 1 (BACE1) expression in the APPswe/PS1dE9 mice. We also found that CCL3 antibody treatment alleviated neuroinflammation and reduced synaptic defects in the APPswe/PS1dE9 mice. Furthermore, the activated NF-κB signaling pathway in APPswe/PS1dE9 mice was inhibited by CCL3 antibody treatment.
CONCLUSIONS: Collectively, our findings provide evidence that CCL3 activation may contribute to the AD pathogenesis and may serve as a novel therapeutic target in the treatment of AD.
Additional Links: PMID-40268067
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40268067,
year = {2025},
author = {Wei, C and Liu, J and Wu, B and Shen, T and Fan, J and Lin, Y and Li, K and Guo, Y and Shang, Y and Zhou, B and Xie, H},
title = {Blockage of CCL3 with neutralizing antibody reduces neuroinflammation and reverses Alzheimer disease phenotypes.},
journal = {Brain, behavior, and immunity},
volume = {},
number = {},
pages = {},
doi = {10.1016/j.bbi.2025.04.034},
pmid = {40268067},
issn = {1090-2139},
abstract = {BACKGROUND: Accumulating evidence indicates that neuroinflammation is involved in the pathogenesis of Alzheimer's disease (AD). According to RNA sequencing and quantitative PCR (qPCR), we found that chemokine CCL3 mRNA expression was abnormally upregulated in the brains of AD transgenic mice. Moreover, the levels of CCL3 in the serum of AD patients were significantly elevated and negatively correlated with their cognitive abilities. However, the role of CCL3 in AD neuroinflammation and pathological damages remains elusive.
METHODS: Using behavioral, histological, and biochemical methods, outcomes of CCL3 antibody treatment on neuropathology and cognitive deficits were studied in the APPswe/PS1dE9 mice.
RESULTS: In the present study, we reported that CCL3 protein expression was increased in the APPswe/PS1dE9 mice, whereas blockage of CCL3 with neutralizing antibody potently inhibited CCL3 activation in the APPswe/PS1dE9 mice down to the levels of wild-type mice. Specifically, CCL3 antibody significantly improved the learning and memory abilities of APPswe/PS1dE9 mice. In addition, CCL3 antibody treatment decreased cerebral amyloid-β (Aβ) levels and plaque burden via inhibiting amyloid precursor protein (APP) processing by reducing beta-site APP cleaving enzyme 1 (BACE1) expression in the APPswe/PS1dE9 mice. We also found that CCL3 antibody treatment alleviated neuroinflammation and reduced synaptic defects in the APPswe/PS1dE9 mice. Furthermore, the activated NF-κB signaling pathway in APPswe/PS1dE9 mice was inhibited by CCL3 antibody treatment.
CONCLUSIONS: Collectively, our findings provide evidence that CCL3 activation may contribute to the AD pathogenesis and may serve as a novel therapeutic target in the treatment of AD.},
}
RevDate: 2025-04-23
Generation of human induced pluripotent stem cell-derived cortical neurons expressing the six tau isoforms.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
BackgroundThe alternative splicing (AS) of MAPT, which encodes Tau, in the adult human brain produces six major isoforms that play critical roles in the pathogenesis of tauopathies including Alzheimer's disease. Previous efforts have failed to differentiate human induced pluripotent stem cells (hiPSCs) to cortical neurons expressing the six isoforms of Tau.ObjectiveWe aim to develop a differentiation method capable of producing the six Tau isoforms in hiPSC-derived cortical neurons.MethodsWe searched for the optimal concentration, duration and treatment window of morphogens in the differentiation of hiPSCs through embryoid bodies (EBs) to dorsal forebrain neuroepithelial cells then to cortical neurons.ResultsThe combined inhibition of WNT, SHH, and SMAD signaling in EBs generated neuroepithelial cells expressing appropriate dorsal forebrain markers, while suppressing ventral, midbrain, and hindbrain genes. Further differentiation in neurogenic and neurotrophic factors produced MAP2[+] neurons at day 18. The iPSC-derived neurons expressed markers of all cortical layers and exhibited synapse formation and synaptic physiology. In addition, MAP2[+] neurons and mitotic cells expressing radial glial markers formed aggregates that could be dissociated to produce mature neurons with similar properties. Most importantly, the six Tau isoforms were expressed from day 80 in a developmentally regulated manner, modeling the situation in human brains on an accelerated timeline.ConclusionsThis chemically defined differentiation method produces a key hallmark of mature human cortical neurons by expressing the six main splicing isoforms of Tau. It will greatly facilitate disease modeling and therapeutic discovery for many human brain disorders involving cortical neurons.
Additional Links: PMID-40267294
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40267294,
year = {2025},
author = {Jiang, H and Xiao, Z and Saleem, K and Zhong, P and Li, L and Chhetri, G and Li, P and Jiang, Z and Yan, Z and Feng, J},
title = {Generation of human induced pluripotent stem cell-derived cortical neurons expressing the six tau isoforms.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251334831},
doi = {10.1177/13872877251334831},
pmid = {40267294},
issn = {1875-8908},
abstract = {BackgroundThe alternative splicing (AS) of MAPT, which encodes Tau, in the adult human brain produces six major isoforms that play critical roles in the pathogenesis of tauopathies including Alzheimer's disease. Previous efforts have failed to differentiate human induced pluripotent stem cells (hiPSCs) to cortical neurons expressing the six isoforms of Tau.ObjectiveWe aim to develop a differentiation method capable of producing the six Tau isoforms in hiPSC-derived cortical neurons.MethodsWe searched for the optimal concentration, duration and treatment window of morphogens in the differentiation of hiPSCs through embryoid bodies (EBs) to dorsal forebrain neuroepithelial cells then to cortical neurons.ResultsThe combined inhibition of WNT, SHH, and SMAD signaling in EBs generated neuroepithelial cells expressing appropriate dorsal forebrain markers, while suppressing ventral, midbrain, and hindbrain genes. Further differentiation in neurogenic and neurotrophic factors produced MAP2[+] neurons at day 18. The iPSC-derived neurons expressed markers of all cortical layers and exhibited synapse formation and synaptic physiology. In addition, MAP2[+] neurons and mitotic cells expressing radial glial markers formed aggregates that could be dissociated to produce mature neurons with similar properties. Most importantly, the six Tau isoforms were expressed from day 80 in a developmentally regulated manner, modeling the situation in human brains on an accelerated timeline.ConclusionsThis chemically defined differentiation method produces a key hallmark of mature human cortical neurons by expressing the six main splicing isoforms of Tau. It will greatly facilitate disease modeling and therapeutic discovery for many human brain disorders involving cortical neurons.},
}
RevDate: 2025-04-23
Drug-targeted Mendelian randomization analysis combined with transcriptome sequencing to explore the molecular mechanisms associated with cognitive impairment.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
BackgroundCurrent therapies for cognitive impairment, including Alzheimer's disease (AD) and mild cognitive impairment, are limited by a lack of universal treatment and adverse effects associated with polypharmacy. Investigating genetic and molecular mechanisms underlying cognitive decline is critical for the development of targeted therapeutics.ObjectiveTo identify causal genes and potential therapeutic targets for cognitive impairment through integrative genomic analyses.MethodsGenome-wide association study data on cognitive impairment were combined with the expression quantitative trait loci (eQTL) data from the eQTLGen consortium. Mendelian randomization (MR) and colocalization analyses were employed to infer causal relationships. Gene Set Enrichment Analysis and Gene Set Variation Analysis evaluated the pathway and functional differences. Immune cell infiltration patterns and the immunometabolic pathways were assessed, followed by drug target prediction.ResultsMR analysis identified seven gene-eQTL pairs significantly associated with cognitive impairment. SMR colocalization prioritized three key genes: HNMT (histamine metabolism), TNFSF8 (inflammatory signaling), and S1PR5 (sphingolipid signaling). HNMT, TNFSF8, and S1PR5 had 39, 24, and 30 predicted targeted drugs, respectively, including arsenic trioxide, aspirin, and immunomodulators.ConclusionsThis study implicates HNMT, TNFSF8, and S1PR5 as potential therapeutic targets for cognitive impairment. Further validation is required to confirm their clinical relevance.
Additional Links: PMID-40267292
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40267292,
year = {2025},
author = {Wu, X and Yang, Q and Xie, Y and Xia, L and Li, J and An, W and Lu, X},
title = {Drug-targeted Mendelian randomization analysis combined with transcriptome sequencing to explore the molecular mechanisms associated with cognitive impairment.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251335891},
doi = {10.1177/13872877251335891},
pmid = {40267292},
issn = {1875-8908},
abstract = {BackgroundCurrent therapies for cognitive impairment, including Alzheimer's disease (AD) and mild cognitive impairment, are limited by a lack of universal treatment and adverse effects associated with polypharmacy. Investigating genetic and molecular mechanisms underlying cognitive decline is critical for the development of targeted therapeutics.ObjectiveTo identify causal genes and potential therapeutic targets for cognitive impairment through integrative genomic analyses.MethodsGenome-wide association study data on cognitive impairment were combined with the expression quantitative trait loci (eQTL) data from the eQTLGen consortium. Mendelian randomization (MR) and colocalization analyses were employed to infer causal relationships. Gene Set Enrichment Analysis and Gene Set Variation Analysis evaluated the pathway and functional differences. Immune cell infiltration patterns and the immunometabolic pathways were assessed, followed by drug target prediction.ResultsMR analysis identified seven gene-eQTL pairs significantly associated with cognitive impairment. SMR colocalization prioritized three key genes: HNMT (histamine metabolism), TNFSF8 (inflammatory signaling), and S1PR5 (sphingolipid signaling). HNMT, TNFSF8, and S1PR5 had 39, 24, and 30 predicted targeted drugs, respectively, including arsenic trioxide, aspirin, and immunomodulators.ConclusionsThis study implicates HNMT, TNFSF8, and S1PR5 as potential therapeutic targets for cognitive impairment. Further validation is required to confirm their clinical relevance.},
}
RevDate: 2025-04-23
Repeated neuromodulation with low-intensity focused ultrasound in patients with Alzheimer's disease.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
BackgroundLow-intensity focused ultrasound (LIFU), a non-invasive targeted brain stimulation technology, has shown promise for therapeutic applications in Alzheimer's disease (AD) patients. Despite its potential, the implications of repeated LIFU neuromodulation in AD patients remain to be investigated.ObjectiveThis pilot study evaluated the safety and potential to improve cognition and functional connectivity following repeated LIFU treatment in AD patients.MethodsTen early-stage AD patients underwent six sessions of neuronavigation-guided LIFU targeting the left dorsolateral prefrontal cortex (DLPFC) within 2-3 weeks, alongside ongoing standard pharmacotherapy. Neuropsychological assessments and resting-state functional magnetic resonance imaging were performed at baseline and eight weeks post-treatment.ResultsMemory performance (p = 0.02) and functional connectivity between the left DLPFC and both the left perirhinal cortex and left dorsomedial prefrontal cortex (corrected p < 0.05) significantly improved from baseline. Additionally, enhancements in memory performance were positively correlated with increases in functional connectivity of the left DLPFC with the left perirhinal cortex (Kendall's tau = 0.56, p = 0.03). No adverse events were reported during the LIFU treatments or at the subsequent follow-up.ConclusionsLIFU may have the therapeutic potential to enhance both brain network connectivity and memory functions in AD patients. Our results provide a basis for further research, including randomized sham-controlled trials and optimization of stimulation protocols, on LIFU as a supplementary or alternative treatment option for AD.Trial registrationClinical Research Information Service, KCT0008169, Registered on 10 February 2023.
Additional Links: PMID-40267289
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40267289,
year = {2025},
author = {Jeong, H and Kim, D and Na, S and Kim, B and Oh, JK and Choi, EK and Yoon, S and Bikson, M and Chung, YA and Song, IU},
title = {Repeated neuromodulation with low-intensity focused ultrasound in patients with Alzheimer's disease.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251333614},
doi = {10.1177/13872877251333614},
pmid = {40267289},
issn = {1875-8908},
abstract = {BackgroundLow-intensity focused ultrasound (LIFU), a non-invasive targeted brain stimulation technology, has shown promise for therapeutic applications in Alzheimer's disease (AD) patients. Despite its potential, the implications of repeated LIFU neuromodulation in AD patients remain to be investigated.ObjectiveThis pilot study evaluated the safety and potential to improve cognition and functional connectivity following repeated LIFU treatment in AD patients.MethodsTen early-stage AD patients underwent six sessions of neuronavigation-guided LIFU targeting the left dorsolateral prefrontal cortex (DLPFC) within 2-3 weeks, alongside ongoing standard pharmacotherapy. Neuropsychological assessments and resting-state functional magnetic resonance imaging were performed at baseline and eight weeks post-treatment.ResultsMemory performance (p = 0.02) and functional connectivity between the left DLPFC and both the left perirhinal cortex and left dorsomedial prefrontal cortex (corrected p < 0.05) significantly improved from baseline. Additionally, enhancements in memory performance were positively correlated with increases in functional connectivity of the left DLPFC with the left perirhinal cortex (Kendall's tau = 0.56, p = 0.03). No adverse events were reported during the LIFU treatments or at the subsequent follow-up.ConclusionsLIFU may have the therapeutic potential to enhance both brain network connectivity and memory functions in AD patients. Our results provide a basis for further research, including randomized sham-controlled trials and optimization of stimulation protocols, on LIFU as a supplementary or alternative treatment option for AD.Trial registrationClinical Research Information Service, KCT0008169, Registered on 10 February 2023.},
}
RevDate: 2025-04-23
Adverse events associated with lecanemab: A disproportionality analysis of data from the FDA adverse event reporting system.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
BackgroundLecanemab, a monoclonal antibody targeting amyloid-β plaques, is FDA-approved for early Alzheimer's disease (AD) treatment. However, safety data from daily clinical practice is limited.ObjectiveThis study aims to assess the adverse events (AEs) linked to lecanemab using the FDA Adverse Event Reporting System (FAERS) to inform better safety management.MethodsA retrospective pharmacovigilance study was conducted using FAERS data from Q1 2023 to Q2 2024. Disproportionality analysis, including reporting odds ratio (ROR), proportional reporting ratio (PRR), Bayesian confidence propagation neural network (BCPNN), and multi-item gamma Poisson shrinker (MGPS), was applied to evaluate AEs where lecanemab was the primary suspect drug.ResultsFrom Q1 2023 to Q2 2024, 917 AEs related to lecanemab were recorded in the FAERS database, with 67.2% of patients aged between 65 and 85 years and 54.5% involving women. Disproportionality analysis identified significant AEs across 22 organ systems, particularly nervous system and psychiatric disorders. Common AEs included headache, amyloid-related imaging abnormalities, and infusion-related reactions, while sleep-related issues like somnolence, abnormal dreams, and poor-quality sleep were notable. Median onset time was 48 days, with serious outcomes in 14.3% of cases, including 70 hospitalizations and 15 deaths.ConclusionsThis pharmacovigilance analysis confirms known AEs of lecanemab and highlights new safety concerns, particularly its impact on sleep. These findings underscore the importance of ongoing monitoring and research to enhance lecanemab's safety profile in AD treatment. However, due to the limitations of FAERS, our analysis is imperfect in terms of important AEs such as therapy-related brain loss and death.
Additional Links: PMID-40267280
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40267280,
year = {2025},
author = {Li, J and Zhang, F and Eisel, UL},
title = {Adverse events associated with lecanemab: A disproportionality analysis of data from the FDA adverse event reporting system.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251333084},
doi = {10.1177/13872877251333084},
pmid = {40267280},
issn = {1875-8908},
abstract = {BackgroundLecanemab, a monoclonal antibody targeting amyloid-β plaques, is FDA-approved for early Alzheimer's disease (AD) treatment. However, safety data from daily clinical practice is limited.ObjectiveThis study aims to assess the adverse events (AEs) linked to lecanemab using the FDA Adverse Event Reporting System (FAERS) to inform better safety management.MethodsA retrospective pharmacovigilance study was conducted using FAERS data from Q1 2023 to Q2 2024. Disproportionality analysis, including reporting odds ratio (ROR), proportional reporting ratio (PRR), Bayesian confidence propagation neural network (BCPNN), and multi-item gamma Poisson shrinker (MGPS), was applied to evaluate AEs where lecanemab was the primary suspect drug.ResultsFrom Q1 2023 to Q2 2024, 917 AEs related to lecanemab were recorded in the FAERS database, with 67.2% of patients aged between 65 and 85 years and 54.5% involving women. Disproportionality analysis identified significant AEs across 22 organ systems, particularly nervous system and psychiatric disorders. Common AEs included headache, amyloid-related imaging abnormalities, and infusion-related reactions, while sleep-related issues like somnolence, abnormal dreams, and poor-quality sleep were notable. Median onset time was 48 days, with serious outcomes in 14.3% of cases, including 70 hospitalizations and 15 deaths.ConclusionsThis pharmacovigilance analysis confirms known AEs of lecanemab and highlights new safety concerns, particularly its impact on sleep. These findings underscore the importance of ongoing monitoring and research to enhance lecanemab's safety profile in AD treatment. However, due to the limitations of FAERS, our analysis is imperfect in terms of important AEs such as therapy-related brain loss and death.},
}
RevDate: 2025-04-23
CmpDate: 2025-04-23
Loss of intracellular ATP affects axoplasmic viscosity and pathological protein aggregation in mammalian neurons.
Science advances, 11(17):eadq6077.
Neurodegenerative diseases display synaptic deficits, mitochondrial defects, and protein aggregation. We show that intracellular adenosine triphosphate (ATP) regulates axoplasmic viscosity and protein aggregation in mammalian neurons. Decreased intracellular ATP upon mitochondrial inhibition leads to axoterminal cytosol, synaptic vesicles, and active zone component condensation, modulating the functional organization of mouse glutamatergic synapses. Proteins involved in the pathogenesis of Parkinson's disease (PD), Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS) condensed and underwent ATP-dependent liquid phase separation in vitro. Human inducible pluripotent stem cell-derived neurons from patients with PD and ALS displayed reduced axoplasmic fluidity and decreased intracellular ATP. Last, nicotinamide mononucleotide treatment successfully rescued intracellular ATP levels and axoplasmic viscosity in neurons from patients with PD and ALS and reduced TAR DNA-binding protein 43 (TDP-43) aggregation in human motor neurons derived from a patient with ALS. Thus, our data suggest that the hydrotropic activity of ATP contributes to the regulation of neuronal homeostasis under both physiological and pathological conditions.
Additional Links: PMID-40267187
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40267187,
year = {2025},
author = {Guillaud, L and Garanzini, A and Zakhia, S and De la Fuente, S and Dimitrov, D and Boerner, S and Terenzio, M},
title = {Loss of intracellular ATP affects axoplasmic viscosity and pathological protein aggregation in mammalian neurons.},
journal = {Science advances},
volume = {11},
number = {17},
pages = {eadq6077},
doi = {10.1126/sciadv.adq6077},
pmid = {40267187},
issn = {2375-2548},
mesh = {*Adenosine Triphosphate/metabolism ; Humans ; Animals ; *Protein Aggregation, Pathological/metabolism/pathology ; Mice ; *Neurons/metabolism ; Induced Pluripotent Stem Cells/metabolism ; Amyotrophic Lateral Sclerosis/metabolism/pathology ; *Axons/metabolism ; Viscosity ; Mitochondria/metabolism ; Protein Aggregates ; Parkinson Disease/metabolism/pathology ; DNA-Binding Proteins/metabolism ; Alzheimer Disease/metabolism/pathology ; },
abstract = {Neurodegenerative diseases display synaptic deficits, mitochondrial defects, and protein aggregation. We show that intracellular adenosine triphosphate (ATP) regulates axoplasmic viscosity and protein aggregation in mammalian neurons. Decreased intracellular ATP upon mitochondrial inhibition leads to axoterminal cytosol, synaptic vesicles, and active zone component condensation, modulating the functional organization of mouse glutamatergic synapses. Proteins involved in the pathogenesis of Parkinson's disease (PD), Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS) condensed and underwent ATP-dependent liquid phase separation in vitro. Human inducible pluripotent stem cell-derived neurons from patients with PD and ALS displayed reduced axoplasmic fluidity and decreased intracellular ATP. Last, nicotinamide mononucleotide treatment successfully rescued intracellular ATP levels and axoplasmic viscosity in neurons from patients with PD and ALS and reduced TAR DNA-binding protein 43 (TDP-43) aggregation in human motor neurons derived from a patient with ALS. Thus, our data suggest that the hydrotropic activity of ATP contributes to the regulation of neuronal homeostasis under both physiological and pathological conditions.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Adenosine Triphosphate/metabolism
Humans
Animals
*Protein Aggregation, Pathological/metabolism/pathology
Mice
*Neurons/metabolism
Induced Pluripotent Stem Cells/metabolism
Amyotrophic Lateral Sclerosis/metabolism/pathology
*Axons/metabolism
Viscosity
Mitochondria/metabolism
Protein Aggregates
Parkinson Disease/metabolism/pathology
DNA-Binding Proteins/metabolism
Alzheimer Disease/metabolism/pathology
RevDate: 2025-04-23
Virtual reality as a complementary therapy in the rehabilitation of balance and gait disorders in patients with mild cognitive impairment and Alzheimer's disease: Systematic review.
Clinical rehabilitation [Epub ahead of print].
ObjectiveTo analyse the benefits of virtual reality in the management of balance and gait disorders in people with Alzheimer's disease and cognitive impairment.Data sourcesPubMed, PEDro, Cochrane Library, Science Direct, Google Scholar and Epistemonikos.Review methodThis study is a systematic review (PROSPERO Registration number: CRD42023486083). The inclusion criteria were: randomised, cross-sectional, quasi-experimental controlled clinical trials involving patients diagnosed with mild cognitive impairment, dementia and Alzheimer's disease with a score of ≤23 on the MMSE test and age ≥60 years, and interventions conducted with virtual reality and conventional physiotherapy for the treatment of balance and gait disorders. The methodological quality and risk of bias assessment was performed with the PEDro scale.Results12 studies were included in the review (n = 476). Three studies applied virtual reality to both experimental and control groups, six applied virtual reality to the experimental group and conventional physiotherapy to the control, and three investigations applied virtual reality to the experimental group and no treatment to the control group. Virtual reality based rehabilitation significantly improved balance and gait, as well as cognitive level, functionality, postural control and mood of the patients compared to those participants who received conventional physiotherapy or no treatment.ConclusionStudies suggest that interventions based on virtual environments in older adults with early Alzheimer's disease can improve balance and gait impairments, postural control and executive function, delaying the deterioration caused by the disease. Furthermore, this therapy has a positive impact on cognitive and motivational performance in these patients.
Additional Links: PMID-40266568
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40266568,
year = {2025},
author = {Rodríguez-Mansilla, J and Chamizo-Gallego, P and González-Sánchez, B and Garrido-Ardila, EM and Torres-Piles, S and Rodríguez-Mansilla, MJ and De Toro-García, Á and Jiménez-Palomares, M},
title = {Virtual reality as a complementary therapy in the rehabilitation of balance and gait disorders in patients with mild cognitive impairment and Alzheimer's disease: Systematic review.},
journal = {Clinical rehabilitation},
volume = {},
number = {},
pages = {2692155251328619},
doi = {10.1177/02692155251328619},
pmid = {40266568},
issn = {1477-0873},
abstract = {ObjectiveTo analyse the benefits of virtual reality in the management of balance and gait disorders in people with Alzheimer's disease and cognitive impairment.Data sourcesPubMed, PEDro, Cochrane Library, Science Direct, Google Scholar and Epistemonikos.Review methodThis study is a systematic review (PROSPERO Registration number: CRD42023486083). The inclusion criteria were: randomised, cross-sectional, quasi-experimental controlled clinical trials involving patients diagnosed with mild cognitive impairment, dementia and Alzheimer's disease with a score of ≤23 on the MMSE test and age ≥60 years, and interventions conducted with virtual reality and conventional physiotherapy for the treatment of balance and gait disorders. The methodological quality and risk of bias assessment was performed with the PEDro scale.Results12 studies were included in the review (n = 476). Three studies applied virtual reality to both experimental and control groups, six applied virtual reality to the experimental group and conventional physiotherapy to the control, and three investigations applied virtual reality to the experimental group and no treatment to the control group. Virtual reality based rehabilitation significantly improved balance and gait, as well as cognitive level, functionality, postural control and mood of the patients compared to those participants who received conventional physiotherapy or no treatment.ConclusionStudies suggest that interventions based on virtual environments in older adults with early Alzheimer's disease can improve balance and gait impairments, postural control and executive function, delaying the deterioration caused by the disease. Furthermore, this therapy has a positive impact on cognitive and motivational performance in these patients.},
}
RevDate: 2025-04-23
Practical brain MRI guidelines for anti-Aβ antibody treatment in early symptomatic Alzheimer's disease.
Japanese journal of radiology [Epub ahead of print].
PURPOSE: These guidelines aim to support magnetic resonance imaging (MRI) diagnosis in patients receiving anti-amyloid β (Aβ) antibody treatment without restricting treatment eligibility.
MATERIALS AND METHODS: These guidelines were collaboratively established by Japan Radiological Society, The Japanese Society of Neuroradiology, and Japanese Society for Magnetic Resonance in Medicine by reviewing existing literature and the results of clinical trials.
RESULTS: Facility standards should comply with the "Optimal Use Promotion Guidelines" of Japan, and physicians should possess comprehensive knowledge of amyloid-related imaging abnormalities (ARIA) and expertise in brain MRI interpretation. The acquisition of knowledge regarding amyloid-related imaging abnormalities, brain MRI, anti-Aβ antibody introduction, and post-treatment diagnosis are also recommended.
CONCLUSION: These guidelines facilitate the accurate diagnosis and effective management of ARIA; ensure the safe administration of anti-Aβ drugs; and provide a framework for MRI facilities, includes staffing requirements and the use of MRI management systems.
Additional Links: PMID-40266549
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40266549,
year = {2025},
author = {Kakeda, S and Miki, Y and Kudo, K and Mori, H and Tokumaru, AM and Abe, O and Aoki, S and , },
title = {Practical brain MRI guidelines for anti-Aβ antibody treatment in early symptomatic Alzheimer's disease.},
journal = {Japanese journal of radiology},
volume = {},
number = {},
pages = {},
pmid = {40266549},
issn = {1867-108X},
abstract = {PURPOSE: These guidelines aim to support magnetic resonance imaging (MRI) diagnosis in patients receiving anti-amyloid β (Aβ) antibody treatment without restricting treatment eligibility.
MATERIALS AND METHODS: These guidelines were collaboratively established by Japan Radiological Society, The Japanese Society of Neuroradiology, and Japanese Society for Magnetic Resonance in Medicine by reviewing existing literature and the results of clinical trials.
RESULTS: Facility standards should comply with the "Optimal Use Promotion Guidelines" of Japan, and physicians should possess comprehensive knowledge of amyloid-related imaging abnormalities (ARIA) and expertise in brain MRI interpretation. The acquisition of knowledge regarding amyloid-related imaging abnormalities, brain MRI, anti-Aβ antibody introduction, and post-treatment diagnosis are also recommended.
CONCLUSION: These guidelines facilitate the accurate diagnosis and effective management of ARIA; ensure the safe administration of anti-Aβ drugs; and provide a framework for MRI facilities, includes staffing requirements and the use of MRI management systems.},
}
RevDate: 2025-04-23
CmpDate: 2025-04-23
Targeting hypoxia-related pathobiology in Alzheimer's disease: strategies for prevention and treatment.
Molecular biology reports, 52(1):416.
INTRODUCTION: Alzheimer's Disease (AD) is a neurodegenerative condition characterised by cognitive decline and memory impairment. Recent research highlights the important role of hypoxia, a state of insufficient oxygen availability, in exacerbating AD pathogenesis.
MATERIALS AND METHODS: Through the use of a number of different search engines like Scopus, PubMed, Bentham, and Elsevier databases, a literature review was carried out for investigating the role of hypoxia mediated pathobiology in AD. Only peerreviewed articles published in reputable journals in English language were included. Conversely, non-peer-reviewed articles, conference abstracts, and editorials were excluded, along with studies lacking experimental or clinical relevance or those unavailable in full text.
CONCLUSION: Hypoxia exacerbates core pathological features such as oxidative stress, neuroinflammation, mitochondrial dysfunction, amyloid-beta (Aβ) dysregulation, and hyperphosphorylation of tau protein. These interlinked mechanisms establish a self-perpetuating cycle of neuronal damage, accelerating disease progression. Addressing hypoxia as a modifiable risk factor offers potential for both prevention and treatment of AD. Exploring hypoxia and the HIF signalling pathway may help counteract the neuropathological and symptomatic effects of neurodegeneration.
Additional Links: PMID-40266407
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40266407,
year = {2025},
author = {Sharma, V and Verma, R and Singh, TG},
title = {Targeting hypoxia-related pathobiology in Alzheimer's disease: strategies for prevention and treatment.},
journal = {Molecular biology reports},
volume = {52},
number = {1},
pages = {416},
pmid = {40266407},
issn = {1573-4978},
mesh = {*Alzheimer Disease/metabolism/prevention & control/pathology/therapy ; Humans ; *Hypoxia/metabolism/complications ; Oxidative Stress ; Amyloid beta-Peptides/metabolism ; Animals ; tau Proteins/metabolism ; Signal Transduction ; Mitochondria/metabolism ; },
abstract = {INTRODUCTION: Alzheimer's Disease (AD) is a neurodegenerative condition characterised by cognitive decline and memory impairment. Recent research highlights the important role of hypoxia, a state of insufficient oxygen availability, in exacerbating AD pathogenesis.
MATERIALS AND METHODS: Through the use of a number of different search engines like Scopus, PubMed, Bentham, and Elsevier databases, a literature review was carried out for investigating the role of hypoxia mediated pathobiology in AD. Only peerreviewed articles published in reputable journals in English language were included. Conversely, non-peer-reviewed articles, conference abstracts, and editorials were excluded, along with studies lacking experimental or clinical relevance or those unavailable in full text.
CONCLUSION: Hypoxia exacerbates core pathological features such as oxidative stress, neuroinflammation, mitochondrial dysfunction, amyloid-beta (Aβ) dysregulation, and hyperphosphorylation of tau protein. These interlinked mechanisms establish a self-perpetuating cycle of neuronal damage, accelerating disease progression. Addressing hypoxia as a modifiable risk factor offers potential for both prevention and treatment of AD. Exploring hypoxia and the HIF signalling pathway may help counteract the neuropathological and symptomatic effects of neurodegeneration.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/metabolism/prevention & control/pathology/therapy
Humans
*Hypoxia/metabolism/complications
Oxidative Stress
Amyloid beta-Peptides/metabolism
Animals
tau Proteins/metabolism
Signal Transduction
Mitochondria/metabolism
RevDate: 2025-04-23
Alzheimer's Disease and Polymeric Nanocarriers: Synergistic Advances in Targeted Drug Delivery.
Current topics in medicinal chemistry pii:CTMC-EPUB-147828 [Epub ahead of print].
INTRODUCTION: Alzheimer's disease (AD) is a prominent neurodegenerative ailment characterized by the constraints of conventional therapies stemming from insufficient medication transport to the brain. This review examines the function of polymeric nanocarriers (PNCs) in improving therapeutic efficacy for Alzheimer's disease treatment.
METHODS: We analyze the principal obstacles to Alzheimer's disease drug delivery: the blood-brain barrier, the blood-cerebrospinal fluid barrier, and multidrug resistance proteins. The review examines three categories of PNCs: polymeric nanoparticles, polymeric micelles, and dendrimers, and their capacity to surmount these obstacles. Literature investigations used search engines like Pub- Med, Google Scholar, and ScienceDirect.
RESULTS: PNCs exhibit superior drug delivery via better biocompatibility, regulated release, and targeted delivery mechanisms. Recent studies demonstrate the effective delivery of several pharmaceuticals, including rivastigmine and galantamine, resulting in enhanced cognitive outcomes in Alzheimer's disease models. Patent research indicates an increase in innovation for PNC-based Alzheimer's disease treatments.
CONCLUSION: Despite ongoing hurdles in biocompatibility and scalability, PNCs exhibit significant potential to transform Alzheimer's disease treatment by improving medication delivery across biological barriers. Current investigations in nanotechnology and combinatorial medicines indicate a favorable outlook for PNC-based medicinal strategies.
Additional Links: PMID-40264328
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40264328,
year = {2025},
author = {Kumar, L and Rana, R and Shaikh, NK and Thakur, A and Kashyap, S and Aggarwal, V and Jyothiraditya, V},
title = {Alzheimer's Disease and Polymeric Nanocarriers: Synergistic Advances in Targeted Drug Delivery.},
journal = {Current topics in medicinal chemistry},
volume = {},
number = {},
pages = {},
doi = {10.2174/0115680266347890250409153450},
pmid = {40264328},
issn = {1873-4294},
abstract = {INTRODUCTION: Alzheimer's disease (AD) is a prominent neurodegenerative ailment characterized by the constraints of conventional therapies stemming from insufficient medication transport to the brain. This review examines the function of polymeric nanocarriers (PNCs) in improving therapeutic efficacy for Alzheimer's disease treatment.
METHODS: We analyze the principal obstacles to Alzheimer's disease drug delivery: the blood-brain barrier, the blood-cerebrospinal fluid barrier, and multidrug resistance proteins. The review examines three categories of PNCs: polymeric nanoparticles, polymeric micelles, and dendrimers, and their capacity to surmount these obstacles. Literature investigations used search engines like Pub- Med, Google Scholar, and ScienceDirect.
RESULTS: PNCs exhibit superior drug delivery via better biocompatibility, regulated release, and targeted delivery mechanisms. Recent studies demonstrate the effective delivery of several pharmaceuticals, including rivastigmine and galantamine, resulting in enhanced cognitive outcomes in Alzheimer's disease models. Patent research indicates an increase in innovation for PNC-based Alzheimer's disease treatments.
CONCLUSION: Despite ongoing hurdles in biocompatibility and scalability, PNCs exhibit significant potential to transform Alzheimer's disease treatment by improving medication delivery across biological barriers. Current investigations in nanotechnology and combinatorial medicines indicate a favorable outlook for PNC-based medicinal strategies.},
}
RevDate: 2025-04-23
Development of Brain Permeable Drugs and Novel Strategies to Overcome the Brain Barriers for Treatment Purposes.
Current pharmaceutical design pii:CPD-EPUB-147800 [Epub ahead of print].
The Blood-Brain Barrier (BBB), a dynamic and highly selective interface, regulates the exchange of molecules between the circulatory system and the Central Nervous System (CNS). While it protects the brain from toxins and pathogens, it also restricts the delivery of therapeutic agents, posing a significant challenge in treating CNS disorders such as Alzheimer's disease, Parkinson's disease, and glioblastoma. This manuscript explores the structural and functional complexity of the BBB, including the roles of tight junctions, adherens junctions, astrocytes, pericytes, and endothelial cells. It highlights the influence of drug physicochemical properties, such as lipophilicity, molecular weight, and hydrogen bonding, on BBB penetration. Current strategies to enhance drug delivery include nanotechnology-based carriers (liposomes, solid lipid nanoparticles, polymer-based carriers), receptor-mediated transcytosis, and cell-penetrating peptides. Emerging approaches like focused ultrasound with microbubbles, intranasal delivery, and exosome-mediated transport demonstrate significant potential for bypassing BBB constraints. Gene therapy, employing both viral and nonviral vectors, offers promise for addressing genetic CNS disorders. Despite advances, limitations, such as offtarget effects, limited delivery efficiency, and potential toxicity, remain critical barriers to clinical translation. Future research must prioritize multidisciplinary approaches integrating nanotechnology, personalized medicine, and enhanced understanding of BBB biology. Innovations in non-invasive, targeted delivery systems are essential to overcoming existing challenges and enabling effective treatment of CNS disorders. This review underscores the need for further exploration of these technologies to achieve sustained, site-specific drug delivery, thereby advancing therapeutic interventions for neurological diseases. Significance Statement: The blood-brain barrier (BBB) is a critical interface that protects the brain but limits drug delivery, posing challenges in treating CNS disorders. Advancing multidisciplinary approaches and innovative delivery systems is essential to overcome these limitations and enable effective therapies for neurological diseases.
Additional Links: PMID-40264324
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40264324,
year = {2025},
author = {Chauhan, A and Jain, S},
title = {Development of Brain Permeable Drugs and Novel Strategies to Overcome the Brain Barriers for Treatment Purposes.},
journal = {Current pharmaceutical design},
volume = {},
number = {},
pages = {},
doi = {10.2174/0113816128386983250410092649},
pmid = {40264324},
issn = {1873-4286},
abstract = {
The Blood-Brain Barrier (BBB), a dynamic and highly selective interface, regulates the exchange of molecules between the circulatory system and the Central Nervous System (CNS). While it protects the brain from toxins and pathogens, it also restricts the delivery of therapeutic agents, posing a significant challenge in treating CNS disorders such as Alzheimer's disease, Parkinson's disease, and glioblastoma. This manuscript explores the structural and functional complexity of the BBB, including the roles of tight junctions, adherens junctions, astrocytes, pericytes, and endothelial cells. It highlights the influence of drug physicochemical properties, such as lipophilicity, molecular weight, and hydrogen bonding, on BBB penetration. Current strategies to enhance drug delivery include nanotechnology-based carriers (liposomes, solid lipid nanoparticles, polymer-based carriers), receptor-mediated transcytosis, and cell-penetrating peptides. Emerging approaches like focused ultrasound with microbubbles, intranasal delivery, and exosome-mediated transport demonstrate significant potential for bypassing BBB constraints. Gene therapy, employing both viral and nonviral vectors, offers promise for addressing genetic CNS disorders. Despite advances, limitations, such as offtarget effects, limited delivery efficiency, and potential toxicity, remain critical barriers to clinical translation. Future research must prioritize multidisciplinary approaches integrating nanotechnology, personalized medicine, and enhanced understanding of BBB biology. Innovations in non-invasive, targeted delivery systems are essential to overcoming existing challenges and enabling effective treatment of CNS disorders. This review underscores the need for further exploration of these technologies to achieve sustained, site-specific drug delivery, thereby advancing therapeutic interventions for neurological diseases. Significance Statement: The blood-brain barrier (BBB) is a critical interface that protects the brain but limits drug delivery, posing challenges in treating CNS disorders. Advancing multidisciplinary approaches and innovative delivery systems is essential to overcome these limitations and enable effective therapies for neurological diseases.},
}
RevDate: 2025-04-23
Mechanistic investigation on compounds from Amorpha fruticosa L. targeting acetylcholinesterase.
Natural product research [Epub ahead of print].
Acetylcholinesterase (AChE) is the enzyme targeted by drugs used for the symptomatic treatment of cognitive decline associated with Alzheimer's disease. While in vitro data suggest the AChE inhibitory potential of A. fruticosa extracts and components such as rotenoids, in-depth mechanistic investigations are missing. A wide array of computational techniques, including ligand-based approaches, molecular docking, molecular dynamics, and machine learning-assisted toxicity prediction were enrolled in the current study, highlighting the rotenoid 6α,12α-dehydrodeguelin as a promising lead for the development of AChE inhibitors.
Additional Links: PMID-40264302
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40264302,
year = {2025},
author = {Durosini, E and Anyanwu, M and Vendrame, T and Gianoncelli, A and Ribaudo, G},
title = {Mechanistic investigation on compounds from Amorpha fruticosa L. targeting acetylcholinesterase.},
journal = {Natural product research},
volume = {},
number = {},
pages = {1-6},
doi = {10.1080/14786419.2025.2495856},
pmid = {40264302},
issn = {1478-6427},
abstract = {Acetylcholinesterase (AChE) is the enzyme targeted by drugs used for the symptomatic treatment of cognitive decline associated with Alzheimer's disease. While in vitro data suggest the AChE inhibitory potential of A. fruticosa extracts and components such as rotenoids, in-depth mechanistic investigations are missing. A wide array of computational techniques, including ligand-based approaches, molecular docking, molecular dynamics, and machine learning-assisted toxicity prediction were enrolled in the current study, highlighting the rotenoid 6α,12α-dehydrodeguelin as a promising lead for the development of AChE inhibitors.},
}
RevDate: 2025-04-23
CmpDate: 2025-04-23
DNA methylation signature of a lifestyle-based resilience index for cognitive health.
Alzheimer's research & therapy, 17(1):88.
Cognitive resilience (CR) contributes to the variability in risk for developing and progressing in Alzheimer's disease (AD) among individuals. Beyond genetics, recent studies highlight the critical role of lifestyle factors in enhancing CR and delaying cognitive decline. DNA methylation (DNAm), an epigenetic mechanism influenced by both genetic and environmental factors, including CR-related lifestyle factors, offers a promising pathway for understanding the biology of CR. We studied DNAm changes associated with the Resilience Index (RI), a composite measure of lifestyle factors, using blood samples from the Healthy Brain Initiative (HBI) cohort. After corrections for multiple comparisons, our analysis identified 19 CpGs and 24 differentially methylated regions significantly associated with the RI, adjusting for covariates age, sex, APOE ε4, and immune cell composition. The RI-associated methylation changes are significantly enriched in pathways related to lipid metabolism, synaptic plasticity, and neuroinflammation, and highlight the connection between cardiovascular health and cognitive function. By identifying RI-associated DNAm, our study provided an alternative approach to discovering future targets and treatment strategies for AD, complementary to the traditional approach of identifying disease-associated variants directly. Furthermore, we developed a Methylation-based Resilience Score (MRS) that successfully predicted future cognitive decline in an external dataset from the Alzheimer's Disease Neuroimaging Initiative (ADNI), even after accounting for age, sex, APOE ε4, years of education, baseline diagnosis, and baseline MMSE score. Our findings are particularly relevant for a better understanding of epigenetic architecture underlying cognitive resilience. Importantly, the significant association between baseline MRS and future cognitive decline demonstrated that DNAm could be a predictive marker for AD, laying the foundation for future studies on personalized AD prevention.
Additional Links: PMID-40264239
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40264239,
year = {2025},
author = {Zhang, W and Lukacsovich, D and Young, JI and Gomez, L and Schmidt, MA and Martin, ER and Kunkle, BW and Chen, XS and O'Shea, DM and Galvin, JE and Wang, L},
title = {DNA methylation signature of a lifestyle-based resilience index for cognitive health.},
journal = {Alzheimer's research & therapy},
volume = {17},
number = {1},
pages = {88},
pmid = {40264239},
issn = {1758-9193},
support = {R01AG062634//US National Institutes of Health/ ; R01AG062634//US National Institutes of Health/ ; R01NS101483//US National Institutes of Health/ ; R61NS135587//US National Institutes of Health/ ; AWD-Neuro TSA-01//University of Miami Team Science Funding Program/ ; AWD-Neuro TSA-01//University of Miami Team Science Funding Program/ ; AWD-Neuro TSA-01//University of Miami Team Science Funding Program/ ; K12TR004555//Miami Clinical and Translational Science Institute/ ; },
mesh = {Humans ; *DNA Methylation ; Male ; Female ; Aged ; *Life Style ; *Resilience, Psychological ; *Cognition/physiology ; Alzheimer Disease/genetics ; Aged, 80 and over ; *Cognitive Dysfunction/genetics ; Cohort Studies ; Epigenesis, Genetic ; },
abstract = {Cognitive resilience (CR) contributes to the variability in risk for developing and progressing in Alzheimer's disease (AD) among individuals. Beyond genetics, recent studies highlight the critical role of lifestyle factors in enhancing CR and delaying cognitive decline. DNA methylation (DNAm), an epigenetic mechanism influenced by both genetic and environmental factors, including CR-related lifestyle factors, offers a promising pathway for understanding the biology of CR. We studied DNAm changes associated with the Resilience Index (RI), a composite measure of lifestyle factors, using blood samples from the Healthy Brain Initiative (HBI) cohort. After corrections for multiple comparisons, our analysis identified 19 CpGs and 24 differentially methylated regions significantly associated with the RI, adjusting for covariates age, sex, APOE ε4, and immune cell composition. The RI-associated methylation changes are significantly enriched in pathways related to lipid metabolism, synaptic plasticity, and neuroinflammation, and highlight the connection between cardiovascular health and cognitive function. By identifying RI-associated DNAm, our study provided an alternative approach to discovering future targets and treatment strategies for AD, complementary to the traditional approach of identifying disease-associated variants directly. Furthermore, we developed a Methylation-based Resilience Score (MRS) that successfully predicted future cognitive decline in an external dataset from the Alzheimer's Disease Neuroimaging Initiative (ADNI), even after accounting for age, sex, APOE ε4, years of education, baseline diagnosis, and baseline MMSE score. Our findings are particularly relevant for a better understanding of epigenetic architecture underlying cognitive resilience. Importantly, the significant association between baseline MRS and future cognitive decline demonstrated that DNAm could be a predictive marker for AD, laying the foundation for future studies on personalized AD prevention.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*DNA Methylation
Male
Female
Aged
*Life Style
*Resilience, Psychological
*Cognition/physiology
Alzheimer Disease/genetics
Aged, 80 and over
*Cognitive Dysfunction/genetics
Cohort Studies
Epigenesis, Genetic
RevDate: 2025-04-23
CmpDate: 2025-04-23
Inhibition of soluble epoxide hydrolase confers neuroprotection and restores microglial homeostasis in a tauopathy mouse model.
Molecular neurodegeneration, 20(1):44.
BACKGROUND: The epoxyeicosatrienoic acids (EETs) are derivatives of the arachidonic acid metabolism with anti-inflammatory activities. However, their efficacy is limited due to the rapid hydrolysis by soluble epoxide hydrolase (sEH). Inhibition of sEH has been shown to stabilize the EETs and reduce neuroinflammation in Aβ mouse models of Alzheimer's disease (AD). However, the role of the sEH-EET signaling pathway in other CNS cell types and neurodegenerative conditions are less understood.
METHODS: Here we investigated the mechanisms and functional role of the sEH-EET axis in tauopathy by treating PS19 mice with a small molecule sEH inhibitor TPPU and by crossing the PS19 mice with Ephx2 (gene encoding sEH) knockout mice. This was followed by single-nucleus RNA-sequencing (snRNA-seq), biochemical and immunohistochemical analysis, and behavioral assessments. Additionally, we examined the effects of the sEH-EET pathway in primary microglia cultures and human induced pluripotent stem cell (iPSC)-derived neurons exhibiting seeding-induced Tau inclusions.
RESULTS: sEH inhibition improved cognitive function, rescued neuronal cell loss, and reduced Tau pathology and microglial reactivity. snRNA-seq revealed that TPPU treatment upregulated genes involved in actin cytoskeleton and excitatory synaptic pathways. Treatment of human iPSC-derived neurons with TPPU enhanced synaptic density without affecting Tau accumulation, suggesting a cell-autonomous neuroprotective effect of sEH blockade. Furthermore, sEH inhibition reversed disease-associated and interferon-responsive microglial states in PS19 mice, while EET supplementation promoted Tau phagocytosis and clearance in primary microglia cultures.
CONCLUSION: These findings demonstrate that sEH blockade or EET augmentation confers therapeutic benefit in neurodegenerative tauopathies by simultaneously targeting neuronal and microglial pathways.
Additional Links: PMID-40264187
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40264187,
year = {2025},
author = {Wang, S and Qi, C and Rajpurohit, C and Ghosh, B and Xiong, W and Wang, B and Qi, Y and Hwang, SH and Hammock, BD and Li, H and Gan, L and Zheng, H},
title = {Inhibition of soluble epoxide hydrolase confers neuroprotection and restores microglial homeostasis in a tauopathy mouse model.},
journal = {Molecular neurodegeneration},
volume = {20},
number = {1},
pages = {44},
pmid = {40264187},
issn = {1750-1326},
support = {093652/NS/NINDS NIH HHS/United States ; 020670/AG/NIA NIH HHS/United States ; 068031/AG/NIA NIH HHS/United States ; 066606/AG/NIA NIH HHS/United States ; Not applicable//Cure Alzheimer's Fund/ ; A25-1690//Harrington Discovery Institute, University Hospitals/ ; },
mesh = {Animals ; *Epoxide Hydrolases/antagonists & inhibitors/metabolism ; *Microglia/drug effects/metabolism ; *Tauopathies/metabolism/drug therapy/pathology ; Mice ; Disease Models, Animal ; *Neuroprotection/drug effects/physiology ; Homeostasis/drug effects/physiology ; Mice, Knockout ; Humans ; Mice, Transgenic ; Neuroprotective Agents/pharmacology ; },
abstract = {BACKGROUND: The epoxyeicosatrienoic acids (EETs) are derivatives of the arachidonic acid metabolism with anti-inflammatory activities. However, their efficacy is limited due to the rapid hydrolysis by soluble epoxide hydrolase (sEH). Inhibition of sEH has been shown to stabilize the EETs and reduce neuroinflammation in Aβ mouse models of Alzheimer's disease (AD). However, the role of the sEH-EET signaling pathway in other CNS cell types and neurodegenerative conditions are less understood.
METHODS: Here we investigated the mechanisms and functional role of the sEH-EET axis in tauopathy by treating PS19 mice with a small molecule sEH inhibitor TPPU and by crossing the PS19 mice with Ephx2 (gene encoding sEH) knockout mice. This was followed by single-nucleus RNA-sequencing (snRNA-seq), biochemical and immunohistochemical analysis, and behavioral assessments. Additionally, we examined the effects of the sEH-EET pathway in primary microglia cultures and human induced pluripotent stem cell (iPSC)-derived neurons exhibiting seeding-induced Tau inclusions.
RESULTS: sEH inhibition improved cognitive function, rescued neuronal cell loss, and reduced Tau pathology and microglial reactivity. snRNA-seq revealed that TPPU treatment upregulated genes involved in actin cytoskeleton and excitatory synaptic pathways. Treatment of human iPSC-derived neurons with TPPU enhanced synaptic density without affecting Tau accumulation, suggesting a cell-autonomous neuroprotective effect of sEH blockade. Furthermore, sEH inhibition reversed disease-associated and interferon-responsive microglial states in PS19 mice, while EET supplementation promoted Tau phagocytosis and clearance in primary microglia cultures.
CONCLUSION: These findings demonstrate that sEH blockade or EET augmentation confers therapeutic benefit in neurodegenerative tauopathies by simultaneously targeting neuronal and microglial pathways.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Epoxide Hydrolases/antagonists & inhibitors/metabolism
*Microglia/drug effects/metabolism
*Tauopathies/metabolism/drug therapy/pathology
Mice
Disease Models, Animal
*Neuroprotection/drug effects/physiology
Homeostasis/drug effects/physiology
Mice, Knockout
Humans
Mice, Transgenic
Neuroprotective Agents/pharmacology
RevDate: 2025-04-22
CmpDate: 2025-04-23
Revolutionizing Alzheimer's disease detection with a cutting-edge CAPCBAM deep learning framework.
Scientific reports, 15(1):13925.
Early and accurate diagnosis of Alzheimer's disease (AD) is crucial for effective treatment. While the integration of deep learning techniques for AD classification is not entirely new, this study introduces CAPCBAM-a framework that extends prior approaches by combining Capsule Networks with a Convolutional Block Attention Module (CBAM). In CAPCBAM, standardized preprocessing of MRI images is followed by feature extraction using Capsule Networks, which preserve spatial hierarchies and capture intricate relationships among image features. The subsequent application of CBAM, employing both channel and spatial attention mechanisms, refines the feature maps to highlight the most clinically relevant regions. This dual-attention strategy offers clear advantages over conventional CNN methods, particularly in enhancing model generalization and mitigating information loss due to pooling. On the ADNI dataset, CAPCBAM achieved an impressive accuracy of 99.95%, with precision and recall both at 99.8%, an AUC of 0.99, and an F1-Score of 99.92%. Although the use of Capsule Networks and attention mechanisms has been explored previously, CAPCBAM distinguishes itself by its robust integration of these components. The study's advantages include improved feature extraction, faster convergence, and superior classification performance, making it a promising tool for the early detection of Alzheimer's disease.
Additional Links: PMID-40263406
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40263406,
year = {2025},
author = {Slimi, H and Abid, S and Sayadi, M},
title = {Revolutionizing Alzheimer's disease detection with a cutting-edge CAPCBAM deep learning framework.},
journal = {Scientific reports},
volume = {15},
number = {1},
pages = {13925},
pmid = {40263406},
issn = {2045-2322},
mesh = {*Alzheimer Disease/diagnosis/diagnostic imaging ; Humans ; *Deep Learning ; Magnetic Resonance Imaging/methods ; Image Processing, Computer-Assisted/methods ; Neural Networks, Computer ; },
abstract = {Early and accurate diagnosis of Alzheimer's disease (AD) is crucial for effective treatment. While the integration of deep learning techniques for AD classification is not entirely new, this study introduces CAPCBAM-a framework that extends prior approaches by combining Capsule Networks with a Convolutional Block Attention Module (CBAM). In CAPCBAM, standardized preprocessing of MRI images is followed by feature extraction using Capsule Networks, which preserve spatial hierarchies and capture intricate relationships among image features. The subsequent application of CBAM, employing both channel and spatial attention mechanisms, refines the feature maps to highlight the most clinically relevant regions. This dual-attention strategy offers clear advantages over conventional CNN methods, particularly in enhancing model generalization and mitigating information loss due to pooling. On the ADNI dataset, CAPCBAM achieved an impressive accuracy of 99.95%, with precision and recall both at 99.8%, an AUC of 0.99, and an F1-Score of 99.92%. Although the use of Capsule Networks and attention mechanisms has been explored previously, CAPCBAM distinguishes itself by its robust integration of these components. The study's advantages include improved feature extraction, faster convergence, and superior classification performance, making it a promising tool for the early detection of Alzheimer's disease.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/diagnosis/diagnostic imaging
Humans
*Deep Learning
Magnetic Resonance Imaging/methods
Image Processing, Computer-Assisted/methods
Neural Networks, Computer
RevDate: 2025-04-22
CmpDate: 2025-04-23
[Electroacupuncture improves synaptic plasticity and cognitive dysfunction via down-regulating HDAC3 in mice of Alzheimer's disease].
Zhen ci yan jiu = Acupuncture research, 50(4):375-383.
OBJECTIVES: To observe the effect of electroacupuncture(EA) on histone deacetylase 3 (HDAC3), synaptic plasticity and N-methyl-D-aspartate (NMDA) receptors in the hippocampus of mice with Alzheimer's disease(AD), so as to explore the underlying mechanism of EA in treatment of AD.
METHODS: 5XFAD mice were randomly divided into EA group, model group and sham-acupuncture group, with 13 mice in both the EA group and the model group, and 7 mice in the sham-acupuncture group. Thirteen wild-type mice from the same litter were taken as the normal control group. The mice in the EA group received EA at "Baihui" (GV20)and "Dazhui" (GV14) for 15 min once daily, 6 times a week for 4 weeks. The mice in the sham-acupuncture group received sham EA, i.e., the needle was inserted into the rubber clay which was placed on the surface of the corresponding acupoints. The novel object recognition(NOR), Y-maze and Morris water maze(MWM) tests were used to observe the cognitive functions of mice. Electrophysiological technique was used to detect long-term potentiation (LTP) of the hippocampal neurons and Western blot was used to detect the relative expressions of HDAC3 and NMDAR-related receptors (NMDAR1, NMDAR2A, NMDAR2B) in the hippocampus.
RESULTS: Compared with the normal control group, 5XFAD mice in the model group showed decreased(P<0.01, P<0.05) preference index for new object recognition, alternative arm ratio (AAR), number of times crossing the original platform, percentage of time and distance traveled in the target quadrant, NMDAR2B, NMDAR2A and NMDAR1 protein expression levels, with prolonged(P<0.01) escape latency, and increased (P<0.05) protein relative expression of HDAC3. At the same time, with high-frequency stimulation, the slope of fEPSP was decreased(P<0.01, P<0.05)in the 5XFAD mice. After EA intervention, comparison between the EA and the model groups revealed that, the preference index for new object recognition, AAR were increased (P<0.01, P<0.05) in the EA group, the escape latency was shortened (P<0.05), and the number of times crossing the platform, percentage of time and distance traveled in the target quadrant, the slope of fEPSP, and the protein relative expressions of NMDAR2B, NMDAR2A and NMDAR1 in the hippocampus were increased (P<0.01, P<0.05), while the protein relative expression of HDAC3 decreased (P<0.01). Compared with sham-acupuncture group, the above indexes improved to different degree in the EA group (P<0.01, P<0.05).
CONCLUSIONS: EA of GV20 and GV14 can restore the impaired LTP and improve the cognitive impairment, which may be related to increasing the expressions of NMDA-related receptor proteins and down-regulating the expression of HDAC3 in the hippocampus of 5XFAD mice.
Additional Links: PMID-40262935
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40262935,
year = {2025},
author = {Lai, LF and Li, X and Li, HZ and Li, ZM and Liu, L and Zhang, YY and Yang, H and Luo, BY and Yi, W and Xu, NG and Zhao, JY},
title = {[Electroacupuncture improves synaptic plasticity and cognitive dysfunction via down-regulating HDAC3 in mice of Alzheimer's disease].},
journal = {Zhen ci yan jiu = Acupuncture research},
volume = {50},
number = {4},
pages = {375-383},
doi = {10.13702/j.1000-0607.20240214},
pmid = {40262935},
issn = {1000-0607},
mesh = {Animals ; *Electroacupuncture ; *Alzheimer Disease/therapy/genetics/enzymology/physiopathology/psychology/metabolism ; Mice ; *Neuronal Plasticity ; *Histone Deacetylases/genetics/metabolism ; Humans ; Male ; *Cognitive Dysfunction/therapy/genetics/enzymology/physiopathology/psychology ; Receptors, N-Methyl-D-Aspartate/metabolism/genetics ; Hippocampus/enzymology/metabolism ; Down-Regulation ; Female ; Acupuncture Points ; },
abstract = {OBJECTIVES: To observe the effect of electroacupuncture(EA) on histone deacetylase 3 (HDAC3), synaptic plasticity and N-methyl-D-aspartate (NMDA) receptors in the hippocampus of mice with Alzheimer's disease(AD), so as to explore the underlying mechanism of EA in treatment of AD.
METHODS: 5XFAD mice were randomly divided into EA group, model group and sham-acupuncture group, with 13 mice in both the EA group and the model group, and 7 mice in the sham-acupuncture group. Thirteen wild-type mice from the same litter were taken as the normal control group. The mice in the EA group received EA at "Baihui" (GV20)and "Dazhui" (GV14) for 15 min once daily, 6 times a week for 4 weeks. The mice in the sham-acupuncture group received sham EA, i.e., the needle was inserted into the rubber clay which was placed on the surface of the corresponding acupoints. The novel object recognition(NOR), Y-maze and Morris water maze(MWM) tests were used to observe the cognitive functions of mice. Electrophysiological technique was used to detect long-term potentiation (LTP) of the hippocampal neurons and Western blot was used to detect the relative expressions of HDAC3 and NMDAR-related receptors (NMDAR1, NMDAR2A, NMDAR2B) in the hippocampus.
RESULTS: Compared with the normal control group, 5XFAD mice in the model group showed decreased(P<0.01, P<0.05) preference index for new object recognition, alternative arm ratio (AAR), number of times crossing the original platform, percentage of time and distance traveled in the target quadrant, NMDAR2B, NMDAR2A and NMDAR1 protein expression levels, with prolonged(P<0.01) escape latency, and increased (P<0.05) protein relative expression of HDAC3. At the same time, with high-frequency stimulation, the slope of fEPSP was decreased(P<0.01, P<0.05)in the 5XFAD mice. After EA intervention, comparison between the EA and the model groups revealed that, the preference index for new object recognition, AAR were increased (P<0.01, P<0.05) in the EA group, the escape latency was shortened (P<0.05), and the number of times crossing the platform, percentage of time and distance traveled in the target quadrant, the slope of fEPSP, and the protein relative expressions of NMDAR2B, NMDAR2A and NMDAR1 in the hippocampus were increased (P<0.01, P<0.05), while the protein relative expression of HDAC3 decreased (P<0.01). Compared with sham-acupuncture group, the above indexes improved to different degree in the EA group (P<0.01, P<0.05).
CONCLUSIONS: EA of GV20 and GV14 can restore the impaired LTP and improve the cognitive impairment, which may be related to increasing the expressions of NMDA-related receptor proteins and down-regulating the expression of HDAC3 in the hippocampus of 5XFAD mice.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Electroacupuncture
*Alzheimer Disease/therapy/genetics/enzymology/physiopathology/psychology/metabolism
Mice
*Neuronal Plasticity
*Histone Deacetylases/genetics/metabolism
Humans
Male
*Cognitive Dysfunction/therapy/genetics/enzymology/physiopathology/psychology
Receptors, N-Methyl-D-Aspartate/metabolism/genetics
Hippocampus/enzymology/metabolism
Down-Regulation
Female
Acupuncture Points
RevDate: 2025-04-22
Development of thermosensitive mucoadhesive gel incorporated lipid microspheres of donepezil for enhanced nose-to-brain delivery.
Journal of biomaterials science. Polymer edition [Epub ahead of print].
Alzheimer's disease (ALZ) is a chronic disease that affects the brain neurons leading to dementia. Donepezil (DPZ), a first-line treatment for ALZ is a potent symptomatic therapeutic agent. However, the oral and transdermal route represents non-targeted delivery, causing various adverse effects. This study presents the successful incorporation of a DPZ-loaded lipid microsphere (DPZ-LM) system into a thermosensitive-mucoadhesive gel (TMG), thereby enhancing the delivery of DPZ through the nose-to-brain route. To optimize the formulations, several evaluations were conducted, resulting in an optimized formulation of LM using Compritol[®] exhibited particle size of 8.75 µm, 98.44% of DPZ entrapped, and 93.40% of DPZ loaded in the system with a sustained release manner in the in vitro studies. The TMG-DPZ-LM was prepared using Pluronic[®] F127 and F68, as the gelling agents, with the addition of sodium alginate, as the mucoadhesive polymer. Following incorporation into TMG-DPZ-LM, the system exhibited excellent physicochemical properties and effective nasal delivery in ex vivo permeation has found that 88.58 ± 12.53 µg/cm[2] and retention studies with a mean concentration of 0.0077 mg of retention DPZ in porcine nasal mucosa. The in vivo pharmacokinetic studies demonstrated that the administration of TMG-DPZ-LM via the nose-to-brain route resulted in a significant (p < 0.05) increase in the Cmax, with 207.24 ± 5.16 µg/cm[3] of DPZ in the brain that exhibited a significantly different profile compared to the other route and formulation. The TMG-DPZ-LM system that was developed in this study was considered to have improved its efficacy in the treatment of ALZ.
Additional Links: PMID-40262568
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40262568,
year = {2025},
author = {Tuna, RW and Achmad, NA and Kurniawan, I and Khairiyah, and Asaf, MB and Sapiun, Z and Himawan, A and Dominguez-Robles, J and Aswad, M and Permana, AD},
title = {Development of thermosensitive mucoadhesive gel incorporated lipid microspheres of donepezil for enhanced nose-to-brain delivery.},
journal = {Journal of biomaterials science. Polymer edition},
volume = {},
number = {},
pages = {1-28},
doi = {10.1080/09205063.2025.2492455},
pmid = {40262568},
issn = {1568-5624},
abstract = {Alzheimer's disease (ALZ) is a chronic disease that affects the brain neurons leading to dementia. Donepezil (DPZ), a first-line treatment for ALZ is a potent symptomatic therapeutic agent. However, the oral and transdermal route represents non-targeted delivery, causing various adverse effects. This study presents the successful incorporation of a DPZ-loaded lipid microsphere (DPZ-LM) system into a thermosensitive-mucoadhesive gel (TMG), thereby enhancing the delivery of DPZ through the nose-to-brain route. To optimize the formulations, several evaluations were conducted, resulting in an optimized formulation of LM using Compritol[®] exhibited particle size of 8.75 µm, 98.44% of DPZ entrapped, and 93.40% of DPZ loaded in the system with a sustained release manner in the in vitro studies. The TMG-DPZ-LM was prepared using Pluronic[®] F127 and F68, as the gelling agents, with the addition of sodium alginate, as the mucoadhesive polymer. Following incorporation into TMG-DPZ-LM, the system exhibited excellent physicochemical properties and effective nasal delivery in ex vivo permeation has found that 88.58 ± 12.53 µg/cm[2] and retention studies with a mean concentration of 0.0077 mg of retention DPZ in porcine nasal mucosa. The in vivo pharmacokinetic studies demonstrated that the administration of TMG-DPZ-LM via the nose-to-brain route resulted in a significant (p < 0.05) increase in the Cmax, with 207.24 ± 5.16 µg/cm[3] of DPZ in the brain that exhibited a significantly different profile compared to the other route and formulation. The TMG-DPZ-LM system that was developed in this study was considered to have improved its efficacy in the treatment of ALZ.},
}
RevDate: 2025-04-22
Resilient together for dementia: A qualitative study of couples' treatment preferences to address distress early after diagnosis.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
BackgroundDespite technological advances and earlier and more confident diagnoses, there is a lack of post-diagnosis support for couples navigating the challenges of early dementia. Clinically elevated emotional distress is common for both partners after diagnosis, and interferes with the health, relationships, and adjustment of both partners if not addressed.ObjectiveOur objective was to gather in-depth information on couples' preferences to inform the development of a proposed dyadic intervention addressing emotional distress early (within 6 months) after one partners' receipt of a dementia diagnosis.MethodsWe recruited couples after a recent dementia diagnosis (N = 16 dyads; 32 participants) from a large academic medical center via direct provider referrals for 60-min virtual dyadic interviews. Data were analyzed using a hybrid inductive-deductive approach to thematic analysis.ResultsWe identified themes within 3 a-priori determined domains. For dyadic intervention format (domain 1), couples preferred to participate in sessions together and to have flexible options for telehealth and in-person participation. Preferences for intervention content (domain 2) included information on dementia, skills to reduce distress and promote resiliency, and support to communicate about the diagnosis and related stress. Barriers and facilitators (domain 3) included denial or hesitation, resource constraints, and interests in learning skills and connecting to others.ConclusionsWe gathered comprehensive information that could be used to adapt existing dyadic interventions and to tailor support to match couples' preferences early after dementia diagnoses. Early interventions should prioritize flexible delivery of information and skills to couples to support adaptive coping following dementia diagnoses.
Additional Links: PMID-40261286
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40261286,
year = {2025},
author = {Bannon, SM and McCage, S and Walker, K and Brewer, J and Ahmad, N and Cornelius, T and Parker, RA and Dams-O'Connor, K and Dickerson, B and Ritchie, CS and Vranceanu, AM},
title = {Resilient together for dementia: A qualitative study of couples' treatment preferences to address distress early after diagnosis.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251332658},
doi = {10.1177/13872877251332658},
pmid = {40261286},
issn = {1875-8908},
abstract = {BackgroundDespite technological advances and earlier and more confident diagnoses, there is a lack of post-diagnosis support for couples navigating the challenges of early dementia. Clinically elevated emotional distress is common for both partners after diagnosis, and interferes with the health, relationships, and adjustment of both partners if not addressed.ObjectiveOur objective was to gather in-depth information on couples' preferences to inform the development of a proposed dyadic intervention addressing emotional distress early (within 6 months) after one partners' receipt of a dementia diagnosis.MethodsWe recruited couples after a recent dementia diagnosis (N = 16 dyads; 32 participants) from a large academic medical center via direct provider referrals for 60-min virtual dyadic interviews. Data were analyzed using a hybrid inductive-deductive approach to thematic analysis.ResultsWe identified themes within 3 a-priori determined domains. For dyadic intervention format (domain 1), couples preferred to participate in sessions together and to have flexible options for telehealth and in-person participation. Preferences for intervention content (domain 2) included information on dementia, skills to reduce distress and promote resiliency, and support to communicate about the diagnosis and related stress. Barriers and facilitators (domain 3) included denial or hesitation, resource constraints, and interests in learning skills and connecting to others.ConclusionsWe gathered comprehensive information that could be used to adapt existing dyadic interventions and to tailor support to match couples' preferences early after dementia diagnoses. Early interventions should prioritize flexible delivery of information and skills to couples to support adaptive coping following dementia diagnoses.},
}
RevDate: 2025-04-22
Clinicians' Perspectives of Twice-Weekly Rivastigmine Patches for Alzheimer's Disease Treatment in Spain: The VIITAL 2S Study.
Patient preference and adherence, 19:1105-1118.
PURPOSE: Administration routes and dosage significantly impact Alzheimer's disease (AD) treatment effectiveness, as compliance in older patients depends on interactions between concomitant treatments, complex dosages, adverse effects, or medication tolerance. This study aims to describe patient and caregiver preferences concerning treatment with rivastigmine twice-weekly transdermal patches from the neurologists' and geriatricians' perspectives.
METHODS: VIITAL-2S was an ecological study based on aggregated data. A total of 250 Spanish neurologists and geriatricians answered a survey on the use, adherence, patient and caregiver satisfaction, and safety (skin tolerability) of twice-weekly rivastigmine patches.
RESULTS: Most participating physicians reported having over 11 years of experience in their specialty. According to their responses, patients with AD attending Neurology and Geriatrics were usually in mild-moderate condition, and a mean of 61.4% received rivastigmine. Around 60% of patients lived with a family member, and over 80% had a caregiver, mainly their partner/spouse or other relative. Of note, more than half of patients attending Neurology and nearly 75% of patients in Geriatrics received 4-10 medications daily. Both specialists recommended the transdermal formulation to patients receiving rivastigmine. In 33.8% and 41.0% of patients receiving daily patches, neurologists and geriatricians, respectively, recommended switching to twice-weekly patches, considering higher administration comfort and caregiver preferences. Physicians reported high/very high satisfaction with twice-weekly patches in nearly 80% of patients. Comparing twice-weekly to daily patches, they observed higher comfort, more caregiver satisfaction, and enhanced adherence. Both specialists manifested preferring twice-weekly rivastigmine patches over daily ones, especially to increase caregivers' comfort, for patients without full-week caregiver support, and in cases of poor compliance with previous treatments.
CONCLUSION: Neurologists and geriatricians consider the twice-weekly rivastigmine patch formulation beneficial for AD treatment in terms of treatment compliance, skin tolerability, satisfaction and comfort for patients and caregivers.
Additional Links: PMID-40260186
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40260186,
year = {2025},
author = {Piñol-Ripoll, G and Salas Carrillo, M and , },
title = {Clinicians' Perspectives of Twice-Weekly Rivastigmine Patches for Alzheimer's Disease Treatment in Spain: The VIITAL 2S Study.},
journal = {Patient preference and adherence},
volume = {19},
number = {},
pages = {1105-1118},
pmid = {40260186},
issn = {1177-889X},
abstract = {PURPOSE: Administration routes and dosage significantly impact Alzheimer's disease (AD) treatment effectiveness, as compliance in older patients depends on interactions between concomitant treatments, complex dosages, adverse effects, or medication tolerance. This study aims to describe patient and caregiver preferences concerning treatment with rivastigmine twice-weekly transdermal patches from the neurologists' and geriatricians' perspectives.
METHODS: VIITAL-2S was an ecological study based on aggregated data. A total of 250 Spanish neurologists and geriatricians answered a survey on the use, adherence, patient and caregiver satisfaction, and safety (skin tolerability) of twice-weekly rivastigmine patches.
RESULTS: Most participating physicians reported having over 11 years of experience in their specialty. According to their responses, patients with AD attending Neurology and Geriatrics were usually in mild-moderate condition, and a mean of 61.4% received rivastigmine. Around 60% of patients lived with a family member, and over 80% had a caregiver, mainly their partner/spouse or other relative. Of note, more than half of patients attending Neurology and nearly 75% of patients in Geriatrics received 4-10 medications daily. Both specialists recommended the transdermal formulation to patients receiving rivastigmine. In 33.8% and 41.0% of patients receiving daily patches, neurologists and geriatricians, respectively, recommended switching to twice-weekly patches, considering higher administration comfort and caregiver preferences. Physicians reported high/very high satisfaction with twice-weekly patches in nearly 80% of patients. Comparing twice-weekly to daily patches, they observed higher comfort, more caregiver satisfaction, and enhanced adherence. Both specialists manifested preferring twice-weekly rivastigmine patches over daily ones, especially to increase caregivers' comfort, for patients without full-week caregiver support, and in cases of poor compliance with previous treatments.
CONCLUSION: Neurologists and geriatricians consider the twice-weekly rivastigmine patch formulation beneficial for AD treatment in terms of treatment compliance, skin tolerability, satisfaction and comfort for patients and caregivers.},
}
RevDate: 2025-04-22
Efficacy of cholinesterase inhibitors treatment in dementia with Lewy bodies: A 3-year follow-up 'real world' study.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
BackgroundDementia with Lewy bodies (DLB) is the second most common dementia after Alzheimer's disease. Currently, no specific therapeutic agents are available for DLB. However, evidence of cholinergic deficits suggests that enhancing central cholinergic function may be a viable therapeutic approach.ObjectiveTo assess cognitive changes in DLB patients treated with cholinesterase inhibitors (ChEIs) in a real-world setting.MethodsThis retrospective study in a prospective database analyzed data from three dementia clinics between May 2012 and December 2022. Patients with DLB were divided into two groups: those treated with ChEIs and those untreated. Differences in changes in multiple cognitive-related scales between the two groups were analyzed.ResultsThe study included 204 DLB patients, with 133 (65.2%) in the ChEIs group and 71 (34.8%) in the non-ChEIs group. Initial demographic and clinical characteristics were similar between groups. Over time, patients in the ChEIs group showed significantly higher scores on the Mini-Mental State Examination and the Montreal Cognitive Assessment compared to the non-ChEIs group, indicating improved cognitive function. No significant differences were observed in activities of daily living scores.ConclusionsChEIs improved cognitive symptoms in DLB patients in the "real world" study. These findings are consistent with those from a previous small-sample randomized controlled trial. Longitudinal data indicate sustained benefits with continuous ChEIs use in three years. Overall, ChEIs show substantial potential for improving cognitive symptoms in DLB patients.
Additional Links: PMID-40259559
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40259559,
year = {2025},
author = {Wu, H and Sun, Z and Gan, J and Wen, C and Shi, Z and Liu, S and Ji, Y},
title = {Efficacy of cholinesterase inhibitors treatment in dementia with Lewy bodies: A 3-year follow-up 'real world' study.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251330902},
doi = {10.1177/13872877251330902},
pmid = {40259559},
issn = {1875-8908},
abstract = {BackgroundDementia with Lewy bodies (DLB) is the second most common dementia after Alzheimer's disease. Currently, no specific therapeutic agents are available for DLB. However, evidence of cholinergic deficits suggests that enhancing central cholinergic function may be a viable therapeutic approach.ObjectiveTo assess cognitive changes in DLB patients treated with cholinesterase inhibitors (ChEIs) in a real-world setting.MethodsThis retrospective study in a prospective database analyzed data from three dementia clinics between May 2012 and December 2022. Patients with DLB were divided into two groups: those treated with ChEIs and those untreated. Differences in changes in multiple cognitive-related scales between the two groups were analyzed.ResultsThe study included 204 DLB patients, with 133 (65.2%) in the ChEIs group and 71 (34.8%) in the non-ChEIs group. Initial demographic and clinical characteristics were similar between groups. Over time, patients in the ChEIs group showed significantly higher scores on the Mini-Mental State Examination and the Montreal Cognitive Assessment compared to the non-ChEIs group, indicating improved cognitive function. No significant differences were observed in activities of daily living scores.ConclusionsChEIs improved cognitive symptoms in DLB patients in the "real world" study. These findings are consistent with those from a previous small-sample randomized controlled trial. Longitudinal data indicate sustained benefits with continuous ChEIs use in three years. Overall, ChEIs show substantial potential for improving cognitive symptoms in DLB patients.},
}
RevDate: 2025-04-21
APOE4 impairs macrophage lipophagy and promotes demyelination of spiral ganglion neurons in mouse cochleae.
Cell death discovery, 11(1):190.
The ApoE-ε4 gene is a well-established genetic risk factor for neurodegenerative diseases, such as Alzheimer's disease and multiple sclerosis, which are characterized by axonal demyelination in the central nervous system. Recent studies have implicated ApoE-ε4 in age-related hearing loss (ARHL), suggesting a potential role of APOE4 isoform in peripheral nervous system degeneration. However, the role of APOE4 in ARHL are still unclear. In this study, we explored the potential role of APOE4 in axonal demyelination of spiral ganglion neurons (SGNs). ApoE-ε4/ε4 (APOE4) and ApoE-ε3/ε3 (APOE3) mice were used to characterize SGNs. The effect of APOE4 on phagocytosis and autophagy as well as intracellular cholesterol level was evaluated in resident cochlear macrophages (RCMs) and mouse bone marrow-derived macrophages (BMDMs). The results showed that significant axonal demyelination was observed in SGNs of 10-month-old APOE4 mice, accompanied by the presence of myelin debris engulfed by RCMs. Meanwhile, inhibited phagocytosis of myelin debris and impaired lipophagy were detected in APOE4 RCMs and APOE4 BMDMs with an aberrant accumulation of lipid droplets (LDs), which could be reversed by trehalose treatment. This study provided a deep insight into the pathogenesis of APOE4-induced axonal demyelination of SGNs associated with the impaired lipophagy in RCMs, which helped to elucidate the underlying mechanism of ApoE-ε4 in ARHL.
Additional Links: PMID-40258814
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40258814,
year = {2025},
author = {Chen, J and Chen, H and Wei, Q and Lu, Y and Wang, T and Pang, X and Xing, G and Chen, Z and Cao, X and Yao, J},
title = {APOE4 impairs macrophage lipophagy and promotes demyelination of spiral ganglion neurons in mouse cochleae.},
journal = {Cell death discovery},
volume = {11},
number = {1},
pages = {190},
pmid = {40258814},
issn = {2058-7716},
support = {82071052//National Natural Science Foundation of China (National Science Foundation of China)/ ; 82200642//National Natural Science Foundation of China (National Science Foundation of China)/ ; 32070587//National Natural Science Foundation of China (National Science Foundation of China)/ ; },
abstract = {The ApoE-ε4 gene is a well-established genetic risk factor for neurodegenerative diseases, such as Alzheimer's disease and multiple sclerosis, which are characterized by axonal demyelination in the central nervous system. Recent studies have implicated ApoE-ε4 in age-related hearing loss (ARHL), suggesting a potential role of APOE4 isoform in peripheral nervous system degeneration. However, the role of APOE4 in ARHL are still unclear. In this study, we explored the potential role of APOE4 in axonal demyelination of spiral ganglion neurons (SGNs). ApoE-ε4/ε4 (APOE4) and ApoE-ε3/ε3 (APOE3) mice were used to characterize SGNs. The effect of APOE4 on phagocytosis and autophagy as well as intracellular cholesterol level was evaluated in resident cochlear macrophages (RCMs) and mouse bone marrow-derived macrophages (BMDMs). The results showed that significant axonal demyelination was observed in SGNs of 10-month-old APOE4 mice, accompanied by the presence of myelin debris engulfed by RCMs. Meanwhile, inhibited phagocytosis of myelin debris and impaired lipophagy were detected in APOE4 RCMs and APOE4 BMDMs with an aberrant accumulation of lipid droplets (LDs), which could be reversed by trehalose treatment. This study provided a deep insight into the pathogenesis of APOE4-induced axonal demyelination of SGNs associated with the impaired lipophagy in RCMs, which helped to elucidate the underlying mechanism of ApoE-ε4 in ARHL.},
}
RevDate: 2025-04-21
CmpDate: 2025-04-21
Transcranial Pulse Stimulation for Alzheimer's Patients.
Journal of visualized experiments : JoVE.
Transcranial pulse stimulation (TPS) is a noninvasive neuromodulation therapy with Conformité Européenne (CE) marking for the treatment of Alzheimer's disease (AD). Initial pilot studies have demonstrated promising effects on cognitive function. This article focuses on the procedure for treating patients with AD using an MRI-guided, neuro-navigated TPS device. The protocol to be followed for this is described in detail, including the necessary procedures and device settings. A brief overview of the representative clinical results published to date is also provided. In addition to significant clinical improvements in cognition and affect, adverse events (AE) and possible adverse device events (ADE) are presented to provide safety data. Finally, the method is critically discussed. In the future, randomized controlled trials should be conducted to rule out any placebo effects. There is also currently a lack of long-term studies with a larger number of patients. Despite these unresolved questions, TPS has the potential as an adjunct treatment for Alzheimer's patients when used in a controlled, scientifically guided setting.
Additional Links: PMID-40257938
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40257938,
year = {2025},
author = {Cont, C and Reinboth, BS and Schütz, C and Stute, N and Galli, A and Schulte, C and Wojtecki, L},
title = {Transcranial Pulse Stimulation for Alzheimer's Patients.},
journal = {Journal of visualized experiments : JoVE},
volume = {},
number = {218},
pages = {},
doi = {10.3791/67176},
pmid = {40257938},
issn = {1940-087X},
mesh = {*Alzheimer Disease/therapy ; Humans ; Magnetic Resonance Imaging/methods ; *Transcranial Direct Current Stimulation/methods/instrumentation ; *Transcranial Magnetic Stimulation/methods/instrumentation ; },
abstract = {Transcranial pulse stimulation (TPS) is a noninvasive neuromodulation therapy with Conformité Européenne (CE) marking for the treatment of Alzheimer's disease (AD). Initial pilot studies have demonstrated promising effects on cognitive function. This article focuses on the procedure for treating patients with AD using an MRI-guided, neuro-navigated TPS device. The protocol to be followed for this is described in detail, including the necessary procedures and device settings. A brief overview of the representative clinical results published to date is also provided. In addition to significant clinical improvements in cognition and affect, adverse events (AE) and possible adverse device events (ADE) are presented to provide safety data. Finally, the method is critically discussed. In the future, randomized controlled trials should be conducted to rule out any placebo effects. There is also currently a lack of long-term studies with a larger number of patients. Despite these unresolved questions, TPS has the potential as an adjunct treatment for Alzheimer's patients when used in a controlled, scientifically guided setting.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/therapy
Humans
Magnetic Resonance Imaging/methods
*Transcranial Direct Current Stimulation/methods/instrumentation
*Transcranial Magnetic Stimulation/methods/instrumentation
RevDate: 2025-04-21
Improved Alzheimer Disease Diagnosis With a Machine Learning Approach and Neuroimaging: Case Study Development.
JMIRx med, 6:e60866 pii:v6i1e60866.
BACKGROUND: Alzheimer disease (AD) is a severe neurological brain disorder. While not curable, earlier detection can help improve symptoms substantially. Machine learning (ML) models are popular and well suited for medical image processing tasks such as computer-aided diagnosis. These techniques can improve the process for an accurate diagnosis of AD.
OBJECTIVE: In this paper, a complete computer-aided diagnosis system for the diagnosis of AD has been presented. We investigate the performance of some of the most used ML techniques for AD detection and classification using neuroimages from the Open Access Series of Imaging Studies (OASIS) and Alzheimer's Disease Neuroimaging Initiative (ADNI) datasets.
METHODS: The system uses artificial neural networks (ANNs) and support vector machines (SVMs) as classifiers, and dimensionality reduction techniques as feature extractors. To retrieve features from the neuroimages, we used principal component analysis (PCA), linear discriminant analysis, and t-distributed stochastic neighbor embedding. These features are fed into feedforward neural networks (FFNNs) and SVM-based ML classifiers. Furthermore, we applied the vision transformer (ViT)-based ANNs in conjunction with data augmentation to distinguish patients with AD from healthy controls.
RESULTS: Experiments were performed on magnetic resonance imaging and positron emission tomography scans. The OASIS dataset included a total of 300 patients, while the ADNI dataset included 231 patients. For OASIS, 90 (30%) patients were healthy and 210 (70%) were severely impaired by AD. Likewise for the ADNI database, a total of 149 (64.5%) patients with AD were detected and 82 (35.5%) patients were used as healthy controls. An important difference was established between healthy patients and patients with AD (P=.02). We examined the effectiveness of the three feature extractors and classifiers using 5-fold cross-validation and confusion matrix-based standard classification metrics, namely, accuracy, sensitivity, specificity, precision, F1-score, and area under the receiver operating characteristic curve (AUROC). Compared with the state-of-the-art performing methods, the success rate was satisfactory for all the created ML models, but SVM and FFNN performed best with the PCA extractor, while the ViT classifier performed best with more data. The data augmentation/ViT approach worked better overall, achieving accuracies of 93.2% (sensitivity=87.2, specificity=90.5, precision=87.6, F1-score=88.7, and AUROC=92) for OASIS and 90.4% (sensitivity=85.4, specificity=88.6, precision=86.9, F1-score=88, and AUROC=90) for ADNI.
CONCLUSIONS: Effective ML models using neuroimaging data could help physicians working on AD diagnosis and will assist them in prescribing timely treatment to patients with AD. Good results were obtained on the OASIS and ADNI datasets with all the proposed classifiers, namely, SVM, FFNN, and ViTs. However, the results show that the ViT model is much better at predicting AD than the other models when a sufficient amount of data are available to perform the training. This highlights that the data augmentation process could impact the overall performance of the ViT model.
Additional Links: PMID-40257754
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40257754,
year = {2025},
author = {Lazli, L},
title = {Improved Alzheimer Disease Diagnosis With a Machine Learning Approach and Neuroimaging: Case Study Development.},
journal = {JMIRx med},
volume = {6},
number = {},
pages = {e60866},
doi = {10.2196/60866},
pmid = {40257754},
issn = {2563-6316},
abstract = {BACKGROUND: Alzheimer disease (AD) is a severe neurological brain disorder. While not curable, earlier detection can help improve symptoms substantially. Machine learning (ML) models are popular and well suited for medical image processing tasks such as computer-aided diagnosis. These techniques can improve the process for an accurate diagnosis of AD.
OBJECTIVE: In this paper, a complete computer-aided diagnosis system for the diagnosis of AD has been presented. We investigate the performance of some of the most used ML techniques for AD detection and classification using neuroimages from the Open Access Series of Imaging Studies (OASIS) and Alzheimer's Disease Neuroimaging Initiative (ADNI) datasets.
METHODS: The system uses artificial neural networks (ANNs) and support vector machines (SVMs) as classifiers, and dimensionality reduction techniques as feature extractors. To retrieve features from the neuroimages, we used principal component analysis (PCA), linear discriminant analysis, and t-distributed stochastic neighbor embedding. These features are fed into feedforward neural networks (FFNNs) and SVM-based ML classifiers. Furthermore, we applied the vision transformer (ViT)-based ANNs in conjunction with data augmentation to distinguish patients with AD from healthy controls.
RESULTS: Experiments were performed on magnetic resonance imaging and positron emission tomography scans. The OASIS dataset included a total of 300 patients, while the ADNI dataset included 231 patients. For OASIS, 90 (30%) patients were healthy and 210 (70%) were severely impaired by AD. Likewise for the ADNI database, a total of 149 (64.5%) patients with AD were detected and 82 (35.5%) patients were used as healthy controls. An important difference was established between healthy patients and patients with AD (P=.02). We examined the effectiveness of the three feature extractors and classifiers using 5-fold cross-validation and confusion matrix-based standard classification metrics, namely, accuracy, sensitivity, specificity, precision, F1-score, and area under the receiver operating characteristic curve (AUROC). Compared with the state-of-the-art performing methods, the success rate was satisfactory for all the created ML models, but SVM and FFNN performed best with the PCA extractor, while the ViT classifier performed best with more data. The data augmentation/ViT approach worked better overall, achieving accuracies of 93.2% (sensitivity=87.2, specificity=90.5, precision=87.6, F1-score=88.7, and AUROC=92) for OASIS and 90.4% (sensitivity=85.4, specificity=88.6, precision=86.9, F1-score=88, and AUROC=90) for ADNI.
CONCLUSIONS: Effective ML models using neuroimaging data could help physicians working on AD diagnosis and will assist them in prescribing timely treatment to patients with AD. Good results were obtained on the OASIS and ADNI datasets with all the proposed classifiers, namely, SVM, FFNN, and ViTs. However, the results show that the ViT model is much better at predicting AD than the other models when a sufficient amount of data are available to perform the training. This highlights that the data augmentation process could impact the overall performance of the ViT model.},
}
RevDate: 2025-04-21
Tailoring MAPK Pathways: New Therapeutic Avenues for Treating Alzheimer's Disease.
Molecular neurobiology [Epub ahead of print].
Alzheimer's disease (AD) is irreversible, progressive, and refractory in nature and is managed very poorly clinically due to very limited treatment outcomes. Unfortunately, most of the multiple clinical trials involving AD patients were unsuccessful in improving the disease prognosis. At the cellular level, many signaling pathways have been proposed to be involved in the sterile/refractory behavior of degenerating neurons in AD. Due to the involvement of p38MAPK in the pathogenesis of Alzheimer's disease, numerous investigations have attempted to determine the beneficial effects of MAPK targeting on memory, inflammatory programming of the brain, and synaptic plasticity. In view of this, various clinical trials involving several MAPK inhibitors (with good safety profiles and few side effects) have yielded positive results in AD patients, suggesting that MAPK targeting may be effective for reducing the pathogenesis of AD, but due to selectivity, dosing, and patient stratification, this aspect still needs further development. In view of their selectivity and off-target effects, only a few MAPK inhibitors have been employed in clinical trials against AD, indicating the scope of their development in this area. Therefore, this study focused on MAPK-based interventions as an upcoming and innovative approach for alleviating AD, with a special emphasis on clinical studies.
Additional Links: PMID-40257689
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40257689,
year = {2025},
author = {Sharma, A and Mehra, V and Kumar, V and Jain, A and Prakash, H},
title = {Tailoring MAPK Pathways: New Therapeutic Avenues for Treating Alzheimer's Disease.},
journal = {Molecular neurobiology},
volume = {},
number = {},
pages = {},
pmid = {40257689},
issn = {1559-1182},
abstract = {Alzheimer's disease (AD) is irreversible, progressive, and refractory in nature and is managed very poorly clinically due to very limited treatment outcomes. Unfortunately, most of the multiple clinical trials involving AD patients were unsuccessful in improving the disease prognosis. At the cellular level, many signaling pathways have been proposed to be involved in the sterile/refractory behavior of degenerating neurons in AD. Due to the involvement of p38MAPK in the pathogenesis of Alzheimer's disease, numerous investigations have attempted to determine the beneficial effects of MAPK targeting on memory, inflammatory programming of the brain, and synaptic plasticity. In view of this, various clinical trials involving several MAPK inhibitors (with good safety profiles and few side effects) have yielded positive results in AD patients, suggesting that MAPK targeting may be effective for reducing the pathogenesis of AD, but due to selectivity, dosing, and patient stratification, this aspect still needs further development. In view of their selectivity and off-target effects, only a few MAPK inhibitors have been employed in clinical trials against AD, indicating the scope of their development in this area. Therefore, this study focused on MAPK-based interventions as an upcoming and innovative approach for alleviating AD, with a special emphasis on clinical studies.},
}
RevDate: 2025-04-21
What Information do Systemic Pathological Changes Bring to the Diagnosis and Treatment of Alzheimer's Disease?.
Neuroscience bulletin [Epub ahead of print].
Alzheimer's disease (AD) is regarded as a neurodegenerative disease, and it has been proposed that AD may be a systemic disease. Studies have reported associations between non-neurological diseases and AD. The correlations between AD pathology and systemic (non-neurological) pathological changes are intricate, and the mechanisms underlying these correlations and their causality are unclear. In this article, we review the association between AD and disorders of other systems. In addition, we summarize the possible mechanisms associated with AD and disorders of other systems, mainly from the perspective of AD pathology. Regarding the relationship between AD and systemic pathological changes, we aim to provide a new outlook on the early warning signs and treatment of AD, such as establishing a diagnostic and screening system based on more accessible peripheral samples.
Additional Links: PMID-40257662
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40257662,
year = {2025},
author = {Zhou, J and Sun, X and Wang, K and Shen, M and Yu, J and Yao, Q and Hong, H and Tang, C and Wang, Q},
title = {What Information do Systemic Pathological Changes Bring to the Diagnosis and Treatment of Alzheimer's Disease?.},
journal = {Neuroscience bulletin},
volume = {},
number = {},
pages = {},
pmid = {40257662},
issn = {1995-8218},
abstract = {Alzheimer's disease (AD) is regarded as a neurodegenerative disease, and it has been proposed that AD may be a systemic disease. Studies have reported associations between non-neurological diseases and AD. The correlations between AD pathology and systemic (non-neurological) pathological changes are intricate, and the mechanisms underlying these correlations and their causality are unclear. In this article, we review the association between AD and disorders of other systems. In addition, we summarize the possible mechanisms associated with AD and disorders of other systems, mainly from the perspective of AD pathology. Regarding the relationship between AD and systemic pathological changes, we aim to provide a new outlook on the early warning signs and treatment of AD, such as establishing a diagnostic and screening system based on more accessible peripheral samples.},
}
RevDate: 2025-04-21
Efficacy of Stem Cell-derived Extracellular Vesicles in the Treatment of Alzheimer's Disease Model Mice: A Systematic Review and Meta-analysis.
Current stem cell research & therapy pii:CSCR-EPUB-147785 [Epub ahead of print].
BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disease that is still incurable. Therapy with stem cell or extracellular vesicles is a promising strategy for AD treatment. Therefore, we evaluated whether stem cell-derived extracellular vesicles could improve cognitive function and pathological features in AD model mice.
METHODS: PubMed, Web of Science, Embase, and The Cochrane Library were searched for studies reporting stem cell-derived extracellular vesicles treatment of AD mice from the establishment of each database to 1st August 2023. SYRCLE was used to assess the risk of bias. The extracted data were analyzed using RevMan 5.4 and Stata 15 software.
RESULTS: 19 studies were included in the analysis. Meta-analysis showed that treatment with stem cell-derived extracellular vesicles significantly improved cognitive performance of AD mice in the Morris water maze test and the novel object recognition test, reduced β-amyloid deposition, alleviated neuroinflammation and decreased levels of the proinflammatory cytokines and glial fibrillary acidic protein (GFAP) in the brain of AD mice. However, stem cell-derived extracellular vesicle did not affect the level of brain phosphorylated tau (p-Tau).
CONCLUSIONS: stem cell-derived extracellular vesicles may promote the degradation of β-amyloid plaques in the brain, regulate immunity and protect nerves, which result in cognitive improvement in AD mice.
Additional Links: PMID-40257023
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40257023,
year = {2025},
author = {Zheng, Q and Wang, S and Wang, T and Zhang, G},
title = {Efficacy of Stem Cell-derived Extracellular Vesicles in the Treatment of Alzheimer's Disease Model Mice: A Systematic Review and Meta-analysis.},
journal = {Current stem cell research & therapy},
volume = {},
number = {},
pages = {},
doi = {10.2174/011574888X352270250407170235},
pmid = {40257023},
issn = {2212-3946},
abstract = {BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disease that is still incurable. Therapy with stem cell or extracellular vesicles is a promising strategy for AD treatment. Therefore, we evaluated whether stem cell-derived extracellular vesicles could improve cognitive function and pathological features in AD model mice.
METHODS: PubMed, Web of Science, Embase, and The Cochrane Library were searched for studies reporting stem cell-derived extracellular vesicles treatment of AD mice from the establishment of each database to 1st August 2023. SYRCLE was used to assess the risk of bias. The extracted data were analyzed using RevMan 5.4 and Stata 15 software.
RESULTS: 19 studies were included in the analysis. Meta-analysis showed that treatment with stem cell-derived extracellular vesicles significantly improved cognitive performance of AD mice in the Morris water maze test and the novel object recognition test, reduced β-amyloid deposition, alleviated neuroinflammation and decreased levels of the proinflammatory cytokines and glial fibrillary acidic protein (GFAP) in the brain of AD mice. However, stem cell-derived extracellular vesicle did not affect the level of brain phosphorylated tau (p-Tau).
CONCLUSIONS: stem cell-derived extracellular vesicles may promote the degradation of β-amyloid plaques in the brain, regulate immunity and protect nerves, which result in cognitive improvement in AD mice.},
}
RevDate: 2025-04-21
County Health Departments Facilitate Telehealth Dementia Evaluation: The Georgia Memory Net Collaboration.
Telemedicine journal and e-health : the official journal of the American Telemedicine Association [Epub ahead of print].
Objective: To establish partnerships with county public health departments to expand telehealth access for referrals to Georgia Memory Net (GMN), a state-funded program focused on improving the early and accurate diagnosis of Alzheimer's disease and other dementias. Method: Funding from the Coronavirus Aid, Relief, and Economic Security Act, and the Building Our Largest Dementia (BOLD) Infrastructure for Alzheimer's Act supported GMN partnership development with Georgia Department of Public Health county health departments. Results: Telehealth infrastructure, including remote video neurological assessments and neuropsychological testing, is now available in 10 Georgia county health departments. Expansion efforts are in progress to bring telehealth services to additional counties, broadening GMN reach. Conclusion: Telehealth evaluations delivered through county health departments provide an effective platform for expanding access to specialized dementia diagnosis and treatment, particularly in rural and underserved areas, enhancing early detection and care for patients throughout the state.
Additional Links: PMID-40256965
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40256965,
year = {2025},
author = {Loring, DW and Lah, JJ and Head, EN and Hale, CL and McIntosh, RL},
title = {County Health Departments Facilitate Telehealth Dementia Evaluation: The Georgia Memory Net Collaboration.},
journal = {Telemedicine journal and e-health : the official journal of the American Telemedicine Association},
volume = {},
number = {},
pages = {},
doi = {10.1089/tmj.2024.0580},
pmid = {40256965},
issn = {1556-3669},
abstract = {Objective: To establish partnerships with county public health departments to expand telehealth access for referrals to Georgia Memory Net (GMN), a state-funded program focused on improving the early and accurate diagnosis of Alzheimer's disease and other dementias. Method: Funding from the Coronavirus Aid, Relief, and Economic Security Act, and the Building Our Largest Dementia (BOLD) Infrastructure for Alzheimer's Act supported GMN partnership development with Georgia Department of Public Health county health departments. Results: Telehealth infrastructure, including remote video neurological assessments and neuropsychological testing, is now available in 10 Georgia county health departments. Expansion efforts are in progress to bring telehealth services to additional counties, broadening GMN reach. Conclusion: Telehealth evaluations delivered through county health departments provide an effective platform for expanding access to specialized dementia diagnosis and treatment, particularly in rural and underserved areas, enhancing early detection and care for patients throughout the state.},
}
RevDate: 2025-04-21
Outer membrane vesicles of Porphyromonas gingivalis: recent advances in pathogenicity and associated mechanisms.
Frontiers in microbiology, 16:1555868.
Periodontitis is a chronic infectious inflammatory disease primarily caused by periodontal pathogenic bacteria, which poses a significant threat to human health. The pathogenic mechanisms associated with Porphyromonas gingivalis (P. gingivalis), a principal causative agent of periodontitis, are particularly complex and warrant thorough investigation. The extensive array of virulence factors released by this bacterium during its growth and pathogenesis not only inflicts localized damage to periodontal tissues but is also intricately linked to the development of systemic diseases through various mechanisms. The outer membrane vesicles (OMVs) produced by P. gingivalis play a key role in this process. These OMVs serve as important mediators of communication between bacteria and host cells and other bacteria, carrying and delivering virulence factors to host cells and distant tissues, thereby damaging host cells and exacerbating inflammatory responses. The ability of these OMVs to disseminate and deliver bacterial virulence factors allows P. gingivalis to play a pathogenic role far beyond the confines of the periodontal tissue and has been closely associated with the development of a variety of systemic diseases such as cardiovascular disease, Alzheimer's disease, rheumatoid arthritis, diabetes mellitus, non-alcoholic hepatitis, and cancer. In view of this, it is of great pathophysiological and clinical significance to deeply investigate its pathogenic role and related mechanisms. This will not only help to better understand the pathogenesis of periodontitis and its related systemic diseases but also provide new ideas and more effective and precise strategies for the early diagnosis, prevention, and treatment of these diseases. However, the current research in this field is still insufficient and in-depth, and many key issues and mechanisms need to be further elucidated. This article summarizes the recent research progress on the role of P. gingivalis OMVs (P. g-OMVs) in related diseases, with the aim of providing a theoretical basis and direction for future research and revealing the pathogenic mechanism of P. g-OMVs more comprehensively.
Additional Links: PMID-40256625
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40256625,
year = {2025},
author = {Wu, Z and Long, W and Yin, Y and Tan, B and Liu, C and Li, H and Ge, S},
title = {Outer membrane vesicles of Porphyromonas gingivalis: recent advances in pathogenicity and associated mechanisms.},
journal = {Frontiers in microbiology},
volume = {16},
number = {},
pages = {1555868},
pmid = {40256625},
issn = {1664-302X},
abstract = {Periodontitis is a chronic infectious inflammatory disease primarily caused by periodontal pathogenic bacteria, which poses a significant threat to human health. The pathogenic mechanisms associated with Porphyromonas gingivalis (P. gingivalis), a principal causative agent of periodontitis, are particularly complex and warrant thorough investigation. The extensive array of virulence factors released by this bacterium during its growth and pathogenesis not only inflicts localized damage to periodontal tissues but is also intricately linked to the development of systemic diseases through various mechanisms. The outer membrane vesicles (OMVs) produced by P. gingivalis play a key role in this process. These OMVs serve as important mediators of communication between bacteria and host cells and other bacteria, carrying and delivering virulence factors to host cells and distant tissues, thereby damaging host cells and exacerbating inflammatory responses. The ability of these OMVs to disseminate and deliver bacterial virulence factors allows P. gingivalis to play a pathogenic role far beyond the confines of the periodontal tissue and has been closely associated with the development of a variety of systemic diseases such as cardiovascular disease, Alzheimer's disease, rheumatoid arthritis, diabetes mellitus, non-alcoholic hepatitis, and cancer. In view of this, it is of great pathophysiological and clinical significance to deeply investigate its pathogenic role and related mechanisms. This will not only help to better understand the pathogenesis of periodontitis and its related systemic diseases but also provide new ideas and more effective and precise strategies for the early diagnosis, prevention, and treatment of these diseases. However, the current research in this field is still insufficient and in-depth, and many key issues and mechanisms need to be further elucidated. This article summarizes the recent research progress on the role of P. gingivalis OMVs (P. g-OMVs) in related diseases, with the aim of providing a theoretical basis and direction for future research and revealing the pathogenic mechanism of P. g-OMVs more comprehensively.},
}
RevDate: 2025-04-21
Targeting ceramide-induced microglial pyroptosis: Icariin is a promising therapy for Alzheimer's disease.
Journal of pharmaceutical analysis, 15(4):101106.
Alzheimer's disease (AD), a progressive dementia, is one of the most common neurodegenerative diseases. Clinical trial results of amyloid-β (Aβ) and tau regulators based on the pretext of straightforward amyloid and tau immunotherapy were disappointing. There are currently no effective strategies for slowing the progression of AD. Herein, we spotlight the dysregulation of lipid metabolism, particularly the elevation of ceramides (Cers), as a critical yet underexplored facet of AD pathogenesis. Our study delineates the role of Cers in promoting microglial pyroptosis, a form of programmed cell death distinct from apoptosis and necroptosis, characterized by cellular swelling, and membrane rupture mediated by the NLRP3 inflammasome pathway. Utilizing both in vivo experiments with amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mice and in vitro assays with BV-2 microglial cells, we investigate the activation of microglial pyroptosis by Cers and its inhibition by icariin (ICA), a flavonoid with known antioxidant and anti-inflammatory properties. Our findings reveal a significant increase in Cers levels and pyroptosis markers (NOD-like receptor family, pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase recruitment domain, caspase-1, gasdermin D (gasdermin D (GSDMD)), and interleukin-18 (IL-18)) in the brains of AD model mice, indicating a direct involvement of Cers in AD pathology through the induction of microglial pyroptosis. Conversely, ICA treatment effectively reduces these pyroptotic markers and Cer levels, thereby attenuating microglial pyroptosis and suggesting a novel therapeutic mechanism of action against AD. This study not only advances our understanding of the pathogenic role of Cers in AD but also introduces ICA as a promising candidate for AD therapy, capable of mitigating neuroinflammation and pyroptosis through the cyclooxygenase-2 (COX-2)-NLRP3 inflammasome-gasdermin D (GSDMD) axis. Our results pave the way for further exploration of Cer metabolism disorders in neurodegenerative diseases and highlight the therapeutic potential of targeting microglial pyroptosis in AD.
Additional Links: PMID-40256246
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40256246,
year = {2025},
author = {Li, H and Xiao, Q and Zhu, L and Kang, J and Zhan, Q and Peng, W},
title = {Targeting ceramide-induced microglial pyroptosis: Icariin is a promising therapy for Alzheimer's disease.},
journal = {Journal of pharmaceutical analysis},
volume = {15},
number = {4},
pages = {101106},
pmid = {40256246},
issn = {2214-0883},
abstract = {Alzheimer's disease (AD), a progressive dementia, is one of the most common neurodegenerative diseases. Clinical trial results of amyloid-β (Aβ) and tau regulators based on the pretext of straightforward amyloid and tau immunotherapy were disappointing. There are currently no effective strategies for slowing the progression of AD. Herein, we spotlight the dysregulation of lipid metabolism, particularly the elevation of ceramides (Cers), as a critical yet underexplored facet of AD pathogenesis. Our study delineates the role of Cers in promoting microglial pyroptosis, a form of programmed cell death distinct from apoptosis and necroptosis, characterized by cellular swelling, and membrane rupture mediated by the NLRP3 inflammasome pathway. Utilizing both in vivo experiments with amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mice and in vitro assays with BV-2 microglial cells, we investigate the activation of microglial pyroptosis by Cers and its inhibition by icariin (ICA), a flavonoid with known antioxidant and anti-inflammatory properties. Our findings reveal a significant increase in Cers levels and pyroptosis markers (NOD-like receptor family, pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase recruitment domain, caspase-1, gasdermin D (gasdermin D (GSDMD)), and interleukin-18 (IL-18)) in the brains of AD model mice, indicating a direct involvement of Cers in AD pathology through the induction of microglial pyroptosis. Conversely, ICA treatment effectively reduces these pyroptotic markers and Cer levels, thereby attenuating microglial pyroptosis and suggesting a novel therapeutic mechanism of action against AD. This study not only advances our understanding of the pathogenic role of Cers in AD but also introduces ICA as a promising candidate for AD therapy, capable of mitigating neuroinflammation and pyroptosis through the cyclooxygenase-2 (COX-2)-NLRP3 inflammasome-gasdermin D (GSDMD) axis. Our results pave the way for further exploration of Cer metabolism disorders in neurodegenerative diseases and highlight the therapeutic potential of targeting microglial pyroptosis in AD.},
}
RevDate: 2025-04-21
An Implantable In-Hydrogel Wireless Supercapacitor-Activated Neuron System Enables Bidirectional Modulation.
Advanced materials (Deerfield Beach, Fla.) [Epub ahead of print].
The bidirectional modulation of cerebral neurons in the brain possesses enhancement and inhibition of neural activity, which is of great interest in the treatment of motor nerve disorders and emotional disorders, and cognitive defects. However, existing approaches usually rely on electrical/electrochemical stimulations, which show low security by implanting metal probes and unidirectional currents with single modulation. Herein, an implantable in-hydrogel wireless supercapacitor-activated neuron system consisting of the coil, diode bridge circuit, in-hydrogel supercapacitor, and stimulation electrodes is fabricated, which provides a bidirectional and adjustable ion diffusion current to safely and effectively excite and inhibit brain neurons. The designed in-hydrogel supercapacitor exhibits a high storage charge ability of ≈90 times larger than the devices without hydrogel encapsulation, owing to the in situ radical addition mechanism. Moreover, the in-hydrogel electrodes are implanted into the thalamus, amygdala, and prefrontal lobes of the brain to evoke the corresponding changes in potential intensity and frequency through the external chargeable coil and diode bridge circuit, which verifies the potential of the multimodule supercapacitor in amelioration and treatment Parkinson's, severe depression, and Alzheimer's disease.
Additional Links: PMID-40255124
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40255124,
year = {2025},
author = {Sheng, X and Du, Z and Gao, Z and Xu, J and Li, L and Shen, G},
title = {An Implantable In-Hydrogel Wireless Supercapacitor-Activated Neuron System Enables Bidirectional Modulation.},
journal = {Advanced materials (Deerfield Beach, Fla.)},
volume = {},
number = {},
pages = {e2504558},
doi = {10.1002/adma.202504558},
pmid = {40255124},
issn = {1521-4095},
abstract = {The bidirectional modulation of cerebral neurons in the brain possesses enhancement and inhibition of neural activity, which is of great interest in the treatment of motor nerve disorders and emotional disorders, and cognitive defects. However, existing approaches usually rely on electrical/electrochemical stimulations, which show low security by implanting metal probes and unidirectional currents with single modulation. Herein, an implantable in-hydrogel wireless supercapacitor-activated neuron system consisting of the coil, diode bridge circuit, in-hydrogel supercapacitor, and stimulation electrodes is fabricated, which provides a bidirectional and adjustable ion diffusion current to safely and effectively excite and inhibit brain neurons. The designed in-hydrogel supercapacitor exhibits a high storage charge ability of ≈90 times larger than the devices without hydrogel encapsulation, owing to the in situ radical addition mechanism. Moreover, the in-hydrogel electrodes are implanted into the thalamus, amygdala, and prefrontal lobes of the brain to evoke the corresponding changes in potential intensity and frequency through the external chargeable coil and diode bridge circuit, which verifies the potential of the multimodule supercapacitor in amelioration and treatment Parkinson's, severe depression, and Alzheimer's disease.},
}
RevDate: 2025-04-21
Key biomarkers in Alzheimer's disease: Insights for diagnosis and treatment strategies.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
Alzheimer's disease (AD) remains a significant global health challenge, characterized by its progressive neurodegeneration and cognitive decline. The urgent need for early diagnosis and effective treatment necessitates the identification of reliable biomarkers that can illuminate the underlying pathophysiology of AD. This review provides a comprehensive overview of the latest advancements in biomarker research, focusing on their applications in diagnosis, prognosis, and therapeutic development. We delve into the multifaceted landscape of AD biomarkers, encompassing molecular, imaging, and fluid-based markers. The integration of these biomarkers, including amyloid-β and tau proteins, neuroimaging modalities, cerebrospinal fluid analysis, and genetic risk factors, offers a more nuanced understanding of AD's complex etiology. By leveraging the power of precision medicine, biomarker-driven approaches can enable personalized treatment strategies and enhance diagnostic accuracy. Moreover, this review highlights the potential of biomarker research to accelerate drug discovery and development. By identifying novel therapeutic targets and monitoring disease progression, biomarkers can facilitate the evaluation of experimental treatments and ultimately improve patient outcomes. In conclusion, this review underscores the critical role of biomarkers in advancing our comprehension of AD and driving the development of effective interventions. By providing a comprehensive overview of the current state-of-the-art, this work aims to inspire future research and contribute to the goal of conquering AD.
Additional Links: PMID-40255041
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40255041,
year = {2025},
author = {Jamal, R and Shaikh, MA and Taleuzzaman, M and Haque, Z and Albratty, M and Alam, MS and Makeen, HA and Zoghebi, K and Saleh, SF},
title = {Key biomarkers in Alzheimer's disease: Insights for diagnosis and treatment strategies.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251330500},
doi = {10.1177/13872877251330500},
pmid = {40255041},
issn = {1875-8908},
abstract = {Alzheimer's disease (AD) remains a significant global health challenge, characterized by its progressive neurodegeneration and cognitive decline. The urgent need for early diagnosis and effective treatment necessitates the identification of reliable biomarkers that can illuminate the underlying pathophysiology of AD. This review provides a comprehensive overview of the latest advancements in biomarker research, focusing on their applications in diagnosis, prognosis, and therapeutic development. We delve into the multifaceted landscape of AD biomarkers, encompassing molecular, imaging, and fluid-based markers. The integration of these biomarkers, including amyloid-β and tau proteins, neuroimaging modalities, cerebrospinal fluid analysis, and genetic risk factors, offers a more nuanced understanding of AD's complex etiology. By leveraging the power of precision medicine, biomarker-driven approaches can enable personalized treatment strategies and enhance diagnostic accuracy. Moreover, this review highlights the potential of biomarker research to accelerate drug discovery and development. By identifying novel therapeutic targets and monitoring disease progression, biomarkers can facilitate the evaluation of experimental treatments and ultimately improve patient outcomes. In conclusion, this review underscores the critical role of biomarkers in advancing our comprehension of AD and driving the development of effective interventions. By providing a comprehensive overview of the current state-of-the-art, this work aims to inspire future research and contribute to the goal of conquering AD.},
}
RevDate: 2025-04-21
Dilemmas in diagnosing Alzheimer's disease: The peril and promise of self-fulfilling prophecies.
Journal of Alzheimer's disease : JAD [Epub ahead of print].
To date, there are limited empirical data to inform various approaches to communication with patients receiving information on Alzheimer's disease (AD) risk or diagnosis during the pre-symptomatic or minimally symptomatic stages. This article explores the ethical implications of psychological responses known as self-fulfilling prophecies that may impact cognitive decline among individuals diagnosed with or at risk for AD. We describe questions these potential effects raise about the ways clinicians communicate with patients, as well as how caregivers may interact with patients. Recent advancements in biomarkers and treatment for AD underscore the urgency of understanding these phenomena and developing appropriate responses.
Additional Links: PMID-40255038
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40255038,
year = {2025},
author = {Malek, J and Levchenko, A and Robinson, JO and Fong, J and Lin, CR and Jackson, GR and Blumenthal-Barby, J and Shulman, JM and McGuire, AL},
title = {Dilemmas in diagnosing Alzheimer's disease: The peril and promise of self-fulfilling prophecies.},
journal = {Journal of Alzheimer's disease : JAD},
volume = {},
number = {},
pages = {13872877251331236},
doi = {10.1177/13872877251331236},
pmid = {40255038},
issn = {1875-8908},
abstract = {To date, there are limited empirical data to inform various approaches to communication with patients receiving information on Alzheimer's disease (AD) risk or diagnosis during the pre-symptomatic or minimally symptomatic stages. This article explores the ethical implications of psychological responses known as self-fulfilling prophecies that may impact cognitive decline among individuals diagnosed with or at risk for AD. We describe questions these potential effects raise about the ways clinicians communicate with patients, as well as how caregivers may interact with patients. Recent advancements in biomarkers and treatment for AD underscore the urgency of understanding these phenomena and developing appropriate responses.},
}
RevDate: 2025-04-21
Modified Cued Recall Test: Longitudinal Analysis of Test Versions and Item Recall in Adults With Down Syndrome.
Journal of intellectual disability research : JIDR [Epub ahead of print].
BACKGROUND: Adults with Down syndrome (DS) have an elevated risk and early age of onset for Alzheimer's disease (AD). To support upcoming clinical AD trials, there is a critical need to establish cognitive outcome measures that can be used to capture intervention effects. One measure that has successfully been used to detect AD-related cognitive decline in the DS population is a measure of episodic memory, the modified Cued Recall Test (mCRT). Demonstrated utility of the mCRT warrants further investigation into comparisons between the A and B versions, free versus cued recall and changes in performance over time to better understand sensitivity for tracking memory decline over time based on age and AD clinical status.
METHOD: Participants were 272 adults with DS aged 25-81 (mean age = 43.12 years, SD = 9.79). Study procedures were completed at three cycles of data collection: baseline, 16-month follow-up and 32-month follow-up. Participants were enrolled in the Alzheimer Biomarker Consortium-Down Syndrome longitudinal study and completed the mCRT as part of a multiday evaluation. Comparisons were made between the A and B versions of the mCRT in recall and intrusion scores. Participants' ratio of free relative to cued recall was also examined at baseline and longitudinally. Participant performance was compared by age group, clinical AD status (cognitively stable [CS], mild cognitive impairment [MCI] or AD dementia) and premorbid level of intellectual disability (ID).
RESULTS: Version differences were identified, with the most salient differences in the moderate and severe/profound ID groups. The mCRT free recall declined with age in CS participants. Free and cued recall scores were lower in those with MCI and AD dementia, with the exception of the mild ID MCI group, whose cued recall scores were not significantly different from the CS group. Decline across 32 months (mCRT total score decline of 1.29 points/year) was observed for CS participants beginning at ≥ 50 years old, with more pronounced declines in adults with DS with an MCI or AD dementia diagnosis (3.36 and 4.20 points/year, respectively).
CONCLUSION: Characterising test version differences and participant free versus cued recall performance on the mCRT is important for understanding performance under testing conditions and to maximise the sensitivity of clinical interventions to capture meaningful effects. Our findings suggest that clinical AD trials for DS should be cautious about using both versions of the mCRT. Examining the profile of free relative to cued recall may enhance sensitivity for detecting treatment benefits for adults with DS across the range of premorbid ID levels.
Additional Links: PMID-40254895
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40254895,
year = {2025},
author = {Schworer, EK and Handen, BL and Krinsky-McHale, S and Hom, CL and Clare, ICH and Harp, JP and Pulsifer, MB and Mapstone, M and Head, E and Christian, BT and Hartley, SL and , },
title = {Modified Cued Recall Test: Longitudinal Analysis of Test Versions and Item Recall in Adults With Down Syndrome.},
journal = {Journal of intellectual disability research : JIDR},
volume = {},
number = {},
pages = {},
doi = {10.1111/jir.13237},
pmid = {40254895},
issn = {1365-2788},
support = {UL1 TR001873/TR/NCATS NIH HHS/United States ; UL1 TR002373/TR/NCATS NIH HHS/United States ; UL1 TR001414/TR/NCATS NIH HHS/United States ; UL1 TR001857/TR/NCATS NIH HHS/United States ; UL1 TR002345/TR/NCATS NIH HHS/United States ; NIHR203312//NIHR Cambridge Biomedical Research Centre/ ; U01 AG051406/AG/NIA NIH HHS/United States ; U01 AG051412/AG/NIA NIH HHS/United States ; U19 AG068054/AG/NIA NIH HHS/United States ; K99AG084738/AG/NIA NIH HHS/United States ; P50 AG008702/AG/NIA NIH HHS/United States ; P30 AG062421/AG/NIA NIH HHS/United States ; P50 AG16537//National Institutes of Health Programs: The Alzheimers Disease Research Centers Program/ ; P50 AG005133/AG/NIA NIH HHS/United States ; P50 AG005681/AG/NIA NIH HHS/United States ; P30 AG062715/AG/NIA NIH HHS/United States ; P30 AG066519/AG/NIA NIH HHS/United States ; U54 HD090256/HD/NICHD NIH HHS/United States ; U54 HD087011/HD/NICHD NIH HHS/United States ; P50 HD105353/HD/NICHD NIH HHS/United States ; U24 AG21886//National Centralized Repository for Alzheimer Disease and Related Dementias/ ; T32HD007489//Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)/ ; //NIHR Applied Research Collaboration East of England/ ; },
abstract = {BACKGROUND: Adults with Down syndrome (DS) have an elevated risk and early age of onset for Alzheimer's disease (AD). To support upcoming clinical AD trials, there is a critical need to establish cognitive outcome measures that can be used to capture intervention effects. One measure that has successfully been used to detect AD-related cognitive decline in the DS population is a measure of episodic memory, the modified Cued Recall Test (mCRT). Demonstrated utility of the mCRT warrants further investigation into comparisons between the A and B versions, free versus cued recall and changes in performance over time to better understand sensitivity for tracking memory decline over time based on age and AD clinical status.
METHOD: Participants were 272 adults with DS aged 25-81 (mean age = 43.12 years, SD = 9.79). Study procedures were completed at three cycles of data collection: baseline, 16-month follow-up and 32-month follow-up. Participants were enrolled in the Alzheimer Biomarker Consortium-Down Syndrome longitudinal study and completed the mCRT as part of a multiday evaluation. Comparisons were made between the A and B versions of the mCRT in recall and intrusion scores. Participants' ratio of free relative to cued recall was also examined at baseline and longitudinally. Participant performance was compared by age group, clinical AD status (cognitively stable [CS], mild cognitive impairment [MCI] or AD dementia) and premorbid level of intellectual disability (ID).
RESULTS: Version differences were identified, with the most salient differences in the moderate and severe/profound ID groups. The mCRT free recall declined with age in CS participants. Free and cued recall scores were lower in those with MCI and AD dementia, with the exception of the mild ID MCI group, whose cued recall scores were not significantly different from the CS group. Decline across 32 months (mCRT total score decline of 1.29 points/year) was observed for CS participants beginning at ≥ 50 years old, with more pronounced declines in adults with DS with an MCI or AD dementia diagnosis (3.36 and 4.20 points/year, respectively).
CONCLUSION: Characterising test version differences and participant free versus cued recall performance on the mCRT is important for understanding performance under testing conditions and to maximise the sensitivity of clinical interventions to capture meaningful effects. Our findings suggest that clinical AD trials for DS should be cautious about using both versions of the mCRT. Examining the profile of free relative to cued recall may enhance sensitivity for detecting treatment benefits for adults with DS across the range of premorbid ID levels.},
}
RevDate: 2025-04-21
Poria cocos Polysaccharide Reshapes Gut Microbiota to Regulate Short-Chain Fatty Acids and Alleviate Neuroinflammation-Related Cognitive Impairment in Alzheimer's Disease.
Journal of agricultural and food chemistry [Epub ahead of print].
Evidence indicates that Poria cocos polysaccharide (PCP) improves cognitive impairment in Alzheimer's disease (AD); however, its underlying mechanism, particularly its relationship with the gut microbiota, remains unclear. In the current study, we aimed to investigate the mechanism of PCP in improving cognitive impairment in AD. The results demonstrated that PCP markedly enhanced cognitive function and mitigated AD-related pathological alterations in 3 × Tg-AD mice. PCP treatment reversed the age-dependent gut microbiota dysbiosis in 3 × Tg-AD mice by 16S rDNA sequencing. The contents of propanoic acid, butanoic acid and isohexanoic acid were increased by short-chain fatty acid determination. In addition, PCP could restore both the intestinal barrier and the blood-brain barrier, as demonstrated by immunofluorescence staining of tight junction proteins. Furthermore, PCP alleviated systemic inflammation and neuroinflammation, as evidenced by reduced LPS levels in circulation and decreased IL-6 levels in the brain, likely by inhibiting the TLR4/NF-κB signaling pathway. In conclusion, PCP can reshape gut microbiota to regulate short-chain fatty acids and alleviate neuroinflammation-related cognitive impairment in AD mice.
Additional Links: PMID-40254847
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40254847,
year = {2025},
author = {Song, M and Zhang, S and Gan, Y and Ding, T and Li, Z and Fan, X},
title = {Poria cocos Polysaccharide Reshapes Gut Microbiota to Regulate Short-Chain Fatty Acids and Alleviate Neuroinflammation-Related Cognitive Impairment in Alzheimer's Disease.},
journal = {Journal of agricultural and food chemistry},
volume = {},
number = {},
pages = {},
doi = {10.1021/acs.jafc.5c01042},
pmid = {40254847},
issn = {1520-5118},
abstract = {Evidence indicates that Poria cocos polysaccharide (PCP) improves cognitive impairment in Alzheimer's disease (AD); however, its underlying mechanism, particularly its relationship with the gut microbiota, remains unclear. In the current study, we aimed to investigate the mechanism of PCP in improving cognitive impairment in AD. The results demonstrated that PCP markedly enhanced cognitive function and mitigated AD-related pathological alterations in 3 × Tg-AD mice. PCP treatment reversed the age-dependent gut microbiota dysbiosis in 3 × Tg-AD mice by 16S rDNA sequencing. The contents of propanoic acid, butanoic acid and isohexanoic acid were increased by short-chain fatty acid determination. In addition, PCP could restore both the intestinal barrier and the blood-brain barrier, as demonstrated by immunofluorescence staining of tight junction proteins. Furthermore, PCP alleviated systemic inflammation and neuroinflammation, as evidenced by reduced LPS levels in circulation and decreased IL-6 levels in the brain, likely by inhibiting the TLR4/NF-κB signaling pathway. In conclusion, PCP can reshape gut microbiota to regulate short-chain fatty acids and alleviate neuroinflammation-related cognitive impairment in AD mice.},
}
RevDate: 2025-04-20
CmpDate: 2025-04-20
Targeting CD38 immunometabolic checkpoint improves metabolic fitness and cognition in a mouse model of Alzheimer's disease.
Nature communications, 16(1):3736.
Protective immunity, essential for brain maintenance and repair, may be compromised in Alzheimer's disease (AD). Here, using high-dimensional single-cell mass cytometry, we find a unique immunometabolic signature in circulating CD4[+] T cells preceding symptom onset in individuals with familial AD, featured by the elevation of CD38 expression. Using female 5xFAD mice, a mouse model of AD, we show that treatment with an antibody directed to CD38 leads to restored metabolic fitness, improved cognitive performance, and attenuated local neuroinflammation. Comprehensive profiling across distinct immunological niches in 5xFAD mice, reveals a high level of disease-associated CD4[+] T cells that produce IL-17A in the dural meninges, previously linked to cognitive decline. Targeting CD38 leads to abrogation of meningeal TH17 immunity and cortical IL-1β, breaking the negative feedback loop between these two compartments. Taken together, the present findings suggest CD38 as an immunometabolic checkpoint that could be adopted as a pre-symptomatic biomarker for early diagnosis of AD, and might also be therapeutically targeted alone or in combination with other immunotherapies for disease modification.
Additional Links: PMID-40254603
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40254603,
year = {2025},
author = {Peralta Ramos, JM and Castellani, G and Kviatcovsky, D and Croese, T and Tsitsou-Kampeli, A and Burgaletto, C and Abellanas, MA and Cahalon, L and Phoebeluc Colaiuta, S and Salame, TM and Kuperman, Y and Savidor, A and Itkin, M and Malitsky, S and Ovadia, S and Ferrera, S and Kalfon, L and Kadmani, S and Samra, N and Paz, R and Rokach, L and Furlan, R and Aharon-Peretz, J and Falik-Zaccai, TC and Schwartz, M},
title = {Targeting CD38 immunometabolic checkpoint improves metabolic fitness and cognition in a mouse model of Alzheimer's disease.},
journal = {Nature communications},
volume = {16},
number = {1},
pages = {3736},
pmid = {40254603},
issn = {2041-1723},
mesh = {Animals ; *Alzheimer Disease/immunology/metabolism/drug therapy ; *ADP-ribosyl Cyclase 1/metabolism/immunology/antagonists & inhibitors ; Disease Models, Animal ; Female ; Mice ; Humans ; Mice, Transgenic ; *Cognition/drug effects ; *Membrane Glycoproteins/metabolism/immunology/antagonists & inhibitors ; CD4-Positive T-Lymphocytes/immunology/metabolism ; Th17 Cells/immunology/metabolism/drug effects ; Interleukin-17/metabolism/immunology ; Male ; Interleukin-1beta/metabolism/immunology ; Meninges/immunology ; },
abstract = {Protective immunity, essential for brain maintenance and repair, may be compromised in Alzheimer's disease (AD). Here, using high-dimensional single-cell mass cytometry, we find a unique immunometabolic signature in circulating CD4[+] T cells preceding symptom onset in individuals with familial AD, featured by the elevation of CD38 expression. Using female 5xFAD mice, a mouse model of AD, we show that treatment with an antibody directed to CD38 leads to restored metabolic fitness, improved cognitive performance, and attenuated local neuroinflammation. Comprehensive profiling across distinct immunological niches in 5xFAD mice, reveals a high level of disease-associated CD4[+] T cells that produce IL-17A in the dural meninges, previously linked to cognitive decline. Targeting CD38 leads to abrogation of meningeal TH17 immunity and cortical IL-1β, breaking the negative feedback loop between these two compartments. Taken together, the present findings suggest CD38 as an immunometabolic checkpoint that could be adopted as a pre-symptomatic biomarker for early diagnosis of AD, and might also be therapeutically targeted alone or in combination with other immunotherapies for disease modification.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Alzheimer Disease/immunology/metabolism/drug therapy
*ADP-ribosyl Cyclase 1/metabolism/immunology/antagonists & inhibitors
Disease Models, Animal
Female
Mice
Humans
Mice, Transgenic
*Cognition/drug effects
*Membrane Glycoproteins/metabolism/immunology/antagonists & inhibitors
CD4-Positive T-Lymphocytes/immunology/metabolism
Th17 Cells/immunology/metabolism/drug effects
Interleukin-17/metabolism/immunology
Male
Interleukin-1beta/metabolism/immunology
Meninges/immunology
RevDate: 2025-04-20
CmpDate: 2025-04-20
JNK Signaling Pathway Activity Alterations in the Rat Hippocampus: Effect of Age, Alzheimer's Disease-Like Pathology Development, and the JNK Inhibitor IQ-1S.
Biochemistry. Biokhimiia, 90(2):265-275.
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder and the leading cause of senile dementia. The key risk factor for a more common (>95% of cases) sporadic form of AD is age. So far, there are no effective methods for AD prevention or treatment. A growing body of evidence indicates that the development of AD and other neurodegenerative diseases is associated with the activation of mitogen-activated protein kinase (MAPK) pathways, and JNK signaling pathway is considered as a potential target for the prevention and treatment of AD. However, the information on alterations in its activity in ontogenesis, which are evaluated by changes in the phosphorylation of its components, is extremely limited. The aim of this study was to compare age-related changes in the activity of JNK signaling pathway in the hippocampus of Wistar rats and senescence-accelerated OXYS rats (which spontaneously develop the key symptoms of AD-like pathology) and to evaluate the effect of the selective JNK3 inhibitor IQ-1S (11H-indeno[1,2-b]quinoxalin-11-one oxime sodium salt). The ability of IQ-1S to suppress accelerated brain aging in OXYS rat has been proven previously, but the effect of this inhibitor on the JNK activity has not been studied. Here, we showed that with age, the activity of the JNK signaling pathway increased in the hippocampus of rats of both strains. At the same time, the manifestation and active progression of AD-like pathology in OXYS rats was accompanied by the increase in the phosphorylation level of the key kinase of this signaling pathway, JNK3, and its target proteins compared to Wistar rats, which allowed us to suggest JNK3 as a potential target for interventions aimed at preventing neurodegenerative processes. This suggestion was supported by the fact that the neuroprotective effect of the selective JNK3 inhibitor IQ-1S and its ability to suppress the development of neurodegenerative processes in OXYS rats were associated with a decrease in the phosphorylation levels of JNK3, c-Jun, APP, and Tau in the hippocampus.
Additional Links: PMID-40254404
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40254404,
year = {2025},
author = {Muraleva, NA and Zhdankina, AA and Khlebnikov, AI and Kolosova, NG},
title = {JNK Signaling Pathway Activity Alterations in the Rat Hippocampus: Effect of Age, Alzheimer's Disease-Like Pathology Development, and the JNK Inhibitor IQ-1S.},
journal = {Biochemistry. Biokhimiia},
volume = {90},
number = {2},
pages = {265-275},
doi = {10.1134/S0006297924603903},
pmid = {40254404},
issn = {1608-3040},
mesh = {Animals ; *Alzheimer Disease/metabolism/pathology/drug therapy ; *Hippocampus/metabolism/pathology/drug effects ; Rats ; Rats, Wistar ; *MAP Kinase Signaling System/drug effects ; Male ; *Aging/metabolism ; *Protein Kinase Inhibitors/pharmacology ; Mitogen-Activated Protein Kinase 10/metabolism/antagonists & inhibitors ; Phosphorylation ; },
abstract = {Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder and the leading cause of senile dementia. The key risk factor for a more common (>95% of cases) sporadic form of AD is age. So far, there are no effective methods for AD prevention or treatment. A growing body of evidence indicates that the development of AD and other neurodegenerative diseases is associated with the activation of mitogen-activated protein kinase (MAPK) pathways, and JNK signaling pathway is considered as a potential target for the prevention and treatment of AD. However, the information on alterations in its activity in ontogenesis, which are evaluated by changes in the phosphorylation of its components, is extremely limited. The aim of this study was to compare age-related changes in the activity of JNK signaling pathway in the hippocampus of Wistar rats and senescence-accelerated OXYS rats (which spontaneously develop the key symptoms of AD-like pathology) and to evaluate the effect of the selective JNK3 inhibitor IQ-1S (11H-indeno[1,2-b]quinoxalin-11-one oxime sodium salt). The ability of IQ-1S to suppress accelerated brain aging in OXYS rat has been proven previously, but the effect of this inhibitor on the JNK activity has not been studied. Here, we showed that with age, the activity of the JNK signaling pathway increased in the hippocampus of rats of both strains. At the same time, the manifestation and active progression of AD-like pathology in OXYS rats was accompanied by the increase in the phosphorylation level of the key kinase of this signaling pathway, JNK3, and its target proteins compared to Wistar rats, which allowed us to suggest JNK3 as a potential target for interventions aimed at preventing neurodegenerative processes. This suggestion was supported by the fact that the neuroprotective effect of the selective JNK3 inhibitor IQ-1S and its ability to suppress the development of neurodegenerative processes in OXYS rats were associated with a decrease in the phosphorylation levels of JNK3, c-Jun, APP, and Tau in the hippocampus.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Alzheimer Disease/metabolism/pathology/drug therapy
*Hippocampus/metabolism/pathology/drug effects
Rats
Rats, Wistar
*MAP Kinase Signaling System/drug effects
Male
*Aging/metabolism
*Protein Kinase Inhibitors/pharmacology
Mitogen-Activated Protein Kinase 10/metabolism/antagonists & inhibitors
Phosphorylation
RevDate: 2025-04-20
Older adults living with Alzheimer's Disease, dementia or mild cognitive impairment with no informal caregiver or care partner: IDEA Café, the first pilot randomized trial intervention for this underserved populations.
Aging & mental health [Epub ahead of print].
OBJECTIVES: This study presents findings from IDEA Café, for older adults aged 50 and older living with early dementia, dementia, Alzheimer's Disease or cognitive impairment (ED/CI) with no informal caregiver or care partner. IDEA Café is a group adaptation of Innovations in Dementia Empowerment and Action (IDEA) (built upon the foundation of RDAD). It was tested with sexual and gender minority (SGM) older adults, as an underserved population.
METHOD: Employing a two-group randomized controlled pilot trial, thirty participants were randomly assigned to IDEA Café (n = 15) or routine medical care (RMC; n = 15). Feasibility and acceptability were assessed. We conducted pre- and post-treatment assessments of primary and secondary outcomes.
RESULTS: IDEA Café was feasible (attendance, participation), acceptable (helpfulness of the program), and met enrollment goals, with 85% of participants reporting treatment as helpful. The treatment group showed significant improvement in physical functioning (p = 0.04), depressive symptomology (p = 0.03), quality of life (p = 0.04), and a reduction in microaggressions (p = 0.05) and social exclusion (p = 0.03). The RMC showed no statistical change from pretest to posttest.
CONCLUSION: A future randomized controlled trial is needed to test the efficacy and sustainability of the intervention and to bring the intervention to scale.
Additional Links: PMID-40253707
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40253707,
year = {2025},
author = {Fredriksen-Goldsen, KI and Kim, HJ and Teri, L and Jones-Cobb, BR and Fazia, D and Petros, R and Berridge, C and Prasad, A and Oswald, A and Emlet, CA},
title = {Older adults living with Alzheimer's Disease, dementia or mild cognitive impairment with no informal caregiver or care partner: IDEA Café, the first pilot randomized trial intervention for this underserved populations.},
journal = {Aging & mental health},
volume = {},
number = {},
pages = {1-9},
doi = {10.1080/13607863.2025.2468893},
pmid = {40253707},
issn = {1364-6915},
abstract = {OBJECTIVES: This study presents findings from IDEA Café, for older adults aged 50 and older living with early dementia, dementia, Alzheimer's Disease or cognitive impairment (ED/CI) with no informal caregiver or care partner. IDEA Café is a group adaptation of Innovations in Dementia Empowerment and Action (IDEA) (built upon the foundation of RDAD). It was tested with sexual and gender minority (SGM) older adults, as an underserved population.
METHOD: Employing a two-group randomized controlled pilot trial, thirty participants were randomly assigned to IDEA Café (n = 15) or routine medical care (RMC; n = 15). Feasibility and acceptability were assessed. We conducted pre- and post-treatment assessments of primary and secondary outcomes.
RESULTS: IDEA Café was feasible (attendance, participation), acceptable (helpfulness of the program), and met enrollment goals, with 85% of participants reporting treatment as helpful. The treatment group showed significant improvement in physical functioning (p = 0.04), depressive symptomology (p = 0.03), quality of life (p = 0.04), and a reduction in microaggressions (p = 0.05) and social exclusion (p = 0.03). The RMC showed no statistical change from pretest to posttest.
CONCLUSION: A future randomized controlled trial is needed to test the efficacy and sustainability of the intervention and to bring the intervention to scale.},
}
RevDate: 2025-04-20
CmpDate: 2025-04-20
Epinephrine as a Therapeutic Agent for Hyperferritinemia in Diabetes Mellitus and Hypertension.
The American journal of case reports, 26:e947289 pii:947289.
BACKGROUND Diabetes mellitus was the first non-communicable disease to be recognized as a 21st century pandemic. Type 2 diabetes (T2DM) results from increased insulin resistance (IR) or relative insulin deficiency. IR impairs glucose disposal, leading to a compensatory hyper-insulinemic state. Increased iron stores as reflected by high serum ferritin (SF) have been associated with the development T2DM and affect glucose homeostasis by impairing tissue response to insulin. Iron overload (IO) is quite common in essential hypertension (HTN). The first clinical effect of epinephrine on SF was reported in 2024, showing that epinephrine resulted in normalization of SF and recovery from severe COVID-19 infection. CASE REPORT A patient with T2DM, HTN, and dyslipidemia associated with hyperferritinemia received the conventional treatment of T2DM and HTN, with a poor control of hyperglycemia and HTN. Since the patient had elevated SF, we obtained informed written consent for epinephrine's use to lower SF. Epinephrine 0.6 mcg/kg was injected subcutaneously under hemodynamic monitoring, and the results showed normalization of SF and complete recovery of T2DM and HTN. CONCLUSIONS Epinephrine can normalize elevated SF by its iron chelating effect; therefore, it can relieve IO and alleviate IR associated with T2DM and HTN. Epinephrine has an anti-inflammatory and scavenging properties that can inhibit ferroptosis. As a new clinical indication, extensive studies are required for further assessment and possible therapeutic uses in IO disorders such as hereditary hemochromatosis (HH), Alzheimer disease (AD), Parkinsonian disease (PD), and multiple sclerosis (MS).
Additional Links: PMID-40253583
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40253583,
year = {2025},
author = {El-Molla, AM and Aboul Fetouh, F and Bawazir, S and Alwahby, YA and Ali, YA and Basseet, AA and Albanna, AH},
title = {Epinephrine as a Therapeutic Agent for Hyperferritinemia in Diabetes Mellitus and Hypertension.},
journal = {The American journal of case reports},
volume = {26},
number = {},
pages = {e947289},
doi = {10.12659/AJCR.947289},
pmid = {40253583},
issn = {1941-5923},
mesh = {Humans ; *Diabetes Mellitus, Type 2/complications/drug therapy ; *Epinephrine/therapeutic use ; *Hypertension/complications/drug therapy ; *Hyperferritinemia/drug therapy/etiology ; Male ; Middle Aged ; Ferritins/blood ; COVID-19 ; },
abstract = {BACKGROUND Diabetes mellitus was the first non-communicable disease to be recognized as a 21st century pandemic. Type 2 diabetes (T2DM) results from increased insulin resistance (IR) or relative insulin deficiency. IR impairs glucose disposal, leading to a compensatory hyper-insulinemic state. Increased iron stores as reflected by high serum ferritin (SF) have been associated with the development T2DM and affect glucose homeostasis by impairing tissue response to insulin. Iron overload (IO) is quite common in essential hypertension (HTN). The first clinical effect of epinephrine on SF was reported in 2024, showing that epinephrine resulted in normalization of SF and recovery from severe COVID-19 infection. CASE REPORT A patient with T2DM, HTN, and dyslipidemia associated with hyperferritinemia received the conventional treatment of T2DM and HTN, with a poor control of hyperglycemia and HTN. Since the patient had elevated SF, we obtained informed written consent for epinephrine's use to lower SF. Epinephrine 0.6 mcg/kg was injected subcutaneously under hemodynamic monitoring, and the results showed normalization of SF and complete recovery of T2DM and HTN. CONCLUSIONS Epinephrine can normalize elevated SF by its iron chelating effect; therefore, it can relieve IO and alleviate IR associated with T2DM and HTN. Epinephrine has an anti-inflammatory and scavenging properties that can inhibit ferroptosis. As a new clinical indication, extensive studies are required for further assessment and possible therapeutic uses in IO disorders such as hereditary hemochromatosis (HH), Alzheimer disease (AD), Parkinsonian disease (PD), and multiple sclerosis (MS).},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Diabetes Mellitus, Type 2/complications/drug therapy
*Epinephrine/therapeutic use
*Hypertension/complications/drug therapy
*Hyperferritinemia/drug therapy/etiology
Male
Middle Aged
Ferritins/blood
COVID-19
RevDate: 2025-04-19
CmpDate: 2025-04-19
Structure-guided screening identified bioactive phytoconstituents Hernandonine and Anolobine as potential inhibitors of dual specificity protein kinase CLK1.
Scientific reports, 15(1):13604.
Neurodegenerative diseases such as Alzheimer's disease (AD) are becoming a global health concern because of their progressive nature and the extent of the disability they cause to the affected individuals. Existing therapies for neurodegenerative diseases are primarily limited to symptomatic management and have shown diminishing efficacy over time, often leading to resistance. Therefore, there is an urgent need for novel therapeutic interventions that offer improved effectiveness with fewer side effects. A promising approach that has been suggested in neurodegenerative diseases, especially AD, is the prevention of tau phosphorylation by using kinase inhibitors. CDC2-like kinase 1 (CLK1) is a dual-specificity proline-directed protein kinase that is involved in both tau phosphorylation and splicing regulation and is thus a potential drug target. Currently, there are several small molecular CLK1 inhibitors, but problems like drug resistance and side effects still exist. In this study, we have used a structure-guided virtual screening approach to screen the bioactive phytoconstituents of Indian medicinal plants for potential CLK1 inhibitors. The systematic virtual screening involving molecular docking, density functional theory (DFT), and pharmacokinetic profiling, pinpointed Hernandonine and Anolobine as promising compounds due to their druglike properties and high binding affinity with CLK1. To assess the stability and interaction of these compounds with CLK1, all-atom molecular dynamics (MD) simulations and essential dynamics were performed for 500 ns. The results indicate that Hernandonine and Anolobine have a high potential for CLK1 inhibition and can be used as promising leads for developing novel effective drugs against neurodegeneration. This work underlines the importance of the combined use of virtual screening and MD simulations in the search for phytochemical-based CLK1 inhibitors, which can lead to the development of new therapeutic approaches in the treatment of neurodegenerative diseases such as AD.
Additional Links: PMID-40253440
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40253440,
year = {2025},
author = {Khan, S and Choudhury, A and Mohammad, T and Shamsi, A and Hassan, MI and Islam, A},
title = {Structure-guided screening identified bioactive phytoconstituents Hernandonine and Anolobine as potential inhibitors of dual specificity protein kinase CLK1.},
journal = {Scientific reports},
volume = {15},
number = {1},
pages = {13604},
pmid = {40253440},
issn = {2045-2322},
mesh = {*Protein Serine-Threonine Kinases/antagonists & inhibitors/chemistry/metabolism ; Molecular Docking Simulation ; *Protein Kinase Inhibitors/chemistry/pharmacology ; *Protein-Tyrosine Kinases/antagonists & inhibitors/chemistry/metabolism ; Humans ; *Phytochemicals/chemistry/pharmacology ; Molecular Dynamics Simulation ; },
abstract = {Neurodegenerative diseases such as Alzheimer's disease (AD) are becoming a global health concern because of their progressive nature and the extent of the disability they cause to the affected individuals. Existing therapies for neurodegenerative diseases are primarily limited to symptomatic management and have shown diminishing efficacy over time, often leading to resistance. Therefore, there is an urgent need for novel therapeutic interventions that offer improved effectiveness with fewer side effects. A promising approach that has been suggested in neurodegenerative diseases, especially AD, is the prevention of tau phosphorylation by using kinase inhibitors. CDC2-like kinase 1 (CLK1) is a dual-specificity proline-directed protein kinase that is involved in both tau phosphorylation and splicing regulation and is thus a potential drug target. Currently, there are several small molecular CLK1 inhibitors, but problems like drug resistance and side effects still exist. In this study, we have used a structure-guided virtual screening approach to screen the bioactive phytoconstituents of Indian medicinal plants for potential CLK1 inhibitors. The systematic virtual screening involving molecular docking, density functional theory (DFT), and pharmacokinetic profiling, pinpointed Hernandonine and Anolobine as promising compounds due to their druglike properties and high binding affinity with CLK1. To assess the stability and interaction of these compounds with CLK1, all-atom molecular dynamics (MD) simulations and essential dynamics were performed for 500 ns. The results indicate that Hernandonine and Anolobine have a high potential for CLK1 inhibition and can be used as promising leads for developing novel effective drugs against neurodegeneration. This work underlines the importance of the combined use of virtual screening and MD simulations in the search for phytochemical-based CLK1 inhibitors, which can lead to the development of new therapeutic approaches in the treatment of neurodegenerative diseases such as AD.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Protein Serine-Threonine Kinases/antagonists & inhibitors/chemistry/metabolism
Molecular Docking Simulation
*Protein Kinase Inhibitors/chemistry/pharmacology
*Protein-Tyrosine Kinases/antagonists & inhibitors/chemistry/metabolism
Humans
*Phytochemicals/chemistry/pharmacology
Molecular Dynamics Simulation
RevDate: 2025-04-19
Challenges and opportunities for novel combination therapies in Alzheimer's disease: a report from the EU/US CTAD Task Force.
The journal of prevention of Alzheimer's disease pii:S2274-5807(25)00108-6 [Epub ahead of print].
Following the recent approvals of anti-amyloid immunotherapies as "first-in-kind" disease-modifying agents for Alzheimer's disease (AD), there is an emerging emphasis in combination therapies, given the complex and multifactorial etiopathogenesis and pathophysiology of the disease. The EU/US CTAD Task Force met in Madrid in October 2024, to discuss biological rationale and methodological issues and outline potential directions for future research in combination therapies. The Task Force agreed on the necessity and urgency of advancing combination therapies for AD treatment. As of January 1, 2024, in the drug development pipeline, there were 21 combination trials (13 % of all trials). The combination of anti-amyloid and anti-tau therapies could become a central focus of the field. Combinations involving anti-inflammatory and immune mechanisms with anti-amyloid or other therapies also have promise. To facilitate the development and implementation of combination therapies, collaborations between sponsors and public-private partnerships are essential. Optimizing the likelihood of success primarily requires leveraging the use of biomarkers and a clearer understanding of the biological mechanisms underpinning AD and their interactions, especially those involving amyloid, tau, and inflammation, that lead to cognitive decline and progression.
Additional Links: PMID-40253240
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40253240,
year = {2025},
author = {Angioni, D and Middleton, L and Bateman, R and Aisen, P and Boxer, A and Sha, S and Zhou, J and Gerlach, I and Raman, R and Fillit, H and Salloway, S and Sperling, R and Vellas, B and Cummings, J},
title = {Challenges and opportunities for novel combination therapies in Alzheimer's disease: a report from the EU/US CTAD Task Force.},
journal = {The journal of prevention of Alzheimer's disease},
volume = {},
number = {},
pages = {100163},
doi = {10.1016/j.tjpad.2025.100163},
pmid = {40253240},
issn = {2426-0266},
abstract = {Following the recent approvals of anti-amyloid immunotherapies as "first-in-kind" disease-modifying agents for Alzheimer's disease (AD), there is an emerging emphasis in combination therapies, given the complex and multifactorial etiopathogenesis and pathophysiology of the disease. The EU/US CTAD Task Force met in Madrid in October 2024, to discuss biological rationale and methodological issues and outline potential directions for future research in combination therapies. The Task Force agreed on the necessity and urgency of advancing combination therapies for AD treatment. As of January 1, 2024, in the drug development pipeline, there were 21 combination trials (13 % of all trials). The combination of anti-amyloid and anti-tau therapies could become a central focus of the field. Combinations involving anti-inflammatory and immune mechanisms with anti-amyloid or other therapies also have promise. To facilitate the development and implementation of combination therapies, collaborations between sponsors and public-private partnerships are essential. Optimizing the likelihood of success primarily requires leveraging the use of biomarkers and a clearer understanding of the biological mechanisms underpinning AD and their interactions, especially those involving amyloid, tau, and inflammation, that lead to cognitive decline and progression.},
}
RevDate: 2025-04-19
Intranasal resveratrol delivery to the brain with chitosan-decorated bovine serum albumin nanoparticles: Advancing Alzheimer's management in old female rats through QbD-based optimization, in vitro evaluation, and in vivo exploration.
International journal of biological macromolecules pii:S0141-8130(25)03852-8 [Epub ahead of print].
This study aims to prepare, characterize, and evaluate the potential of chitosan-coated bovine serum albumin nanoparticles (CS-BSANPs) loaded with resveratrol (RES) to enhance the therapeutic properties of RES and target Alzheimer's disease in elderly females. As confirmed by morphological analysis, the BSANPs were synthesized using desolvation techniques, resulting in spherical and smooth nanoparticles. Both RES-BSANPs and CS-RES-BSANPs exhibited stability for 90 days at ambient refrigerated temperatures. Through optimization using a Box-Behnken design, RES-BSANPs with favorable colloidal properties were achieved. Differential scanning calorimetry and powder X-ray diffraction confirmed the amorphous dispersion of RES within the nanocarriers. In vitro drug release studies demonstrated a biphasic release pattern aligned with the Korsmeyer-Peppas model, exhibiting both burst and sustained release phases. Stability tests indicated that RES-BSA-NPs and CS-RES-NPs remain stable at 4 °C. Ex vivo studies verified the safety of RES-loaded nanoparticles, and behavioral tests on the Wistar rat model showed that intranasally administered CS-RES-BSANPs were more effective than plain RES dispersion. These results emphasize the potential of biodegradable and mucoadhesive CS-RES-BSANPs as effective drug carriers for intranasal delivery to the brain, offering safety and high tolerability for Alzheimer's disease treatment.
Additional Links: PMID-40253039
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40253039,
year = {2025},
author = {Malaiya, A and Kenwat, R and Mamgain, A and Nayak, P and Parker, A and Paliwal, SR and Paliwal, R},
title = {Intranasal resveratrol delivery to the brain with chitosan-decorated bovine serum albumin nanoparticles: Advancing Alzheimer's management in old female rats through QbD-based optimization, in vitro evaluation, and in vivo exploration.},
journal = {International journal of biological macromolecules},
volume = {},
number = {},
pages = {143300},
doi = {10.1016/j.ijbiomac.2025.143300},
pmid = {40253039},
issn = {1879-0003},
abstract = {This study aims to prepare, characterize, and evaluate the potential of chitosan-coated bovine serum albumin nanoparticles (CS-BSANPs) loaded with resveratrol (RES) to enhance the therapeutic properties of RES and target Alzheimer's disease in elderly females. As confirmed by morphological analysis, the BSANPs were synthesized using desolvation techniques, resulting in spherical and smooth nanoparticles. Both RES-BSANPs and CS-RES-BSANPs exhibited stability for 90 days at ambient refrigerated temperatures. Through optimization using a Box-Behnken design, RES-BSANPs with favorable colloidal properties were achieved. Differential scanning calorimetry and powder X-ray diffraction confirmed the amorphous dispersion of RES within the nanocarriers. In vitro drug release studies demonstrated a biphasic release pattern aligned with the Korsmeyer-Peppas model, exhibiting both burst and sustained release phases. Stability tests indicated that RES-BSA-NPs and CS-RES-NPs remain stable at 4 °C. Ex vivo studies verified the safety of RES-loaded nanoparticles, and behavioral tests on the Wistar rat model showed that intranasally administered CS-RES-BSANPs were more effective than plain RES dispersion. These results emphasize the potential of biodegradable and mucoadhesive CS-RES-BSANPs as effective drug carriers for intranasal delivery to the brain, offering safety and high tolerability for Alzheimer's disease treatment.},
}
RevDate: 2025-04-19
Accelerated iTBS-Induced Changes in Resting-State Functional Connectivity Correspond with Cognitive Improvement in Amnestic MCI.
Brain stimulation pii:S1935-861X(25)00093-2 [Epub ahead of print].
BACKGROUND: Published results of our Phase I safety and feasibility trial of accelerated intermittent theta burst stimulation (a-iTBS) in mild cognitive impairment (MCI) due to Alzheimer's disease showed a large effect-size improvement in cognition.
OBJECTIVE: Further demonstrate target engagement by identifying whether changes in local and network-level functional connectivity relate to the observed cognitive improvement.
METHODS: Eighteen patients with MCI received 3-day a-iTBS (8 sessions/day) to the left dorsolateral prefrontal cortex at Beam F3 (14,400 total pulses) and completed MRI and cognitive testing at pre- and post-treatment. Based on electric field models, we selected 3 stimulated target regions of interest (ROIs) which belonged to the frontoparietal (FPN), default mode (DMN), and ventral attention (VAT) networks (3 target networks). Metrics of resting-state functional connectivity were computed at the ROI level (within-network degree: number of connections) and network level (segregation: strength of connectivity within network relative to other networks). We correlated changes in cognition and connectivity for the target ROIs and networks; off-target ROI (primary visual) and networks serve as negative controls.
RESULTS: Improvements in cognition were associated with connectivity changes in the target ROIs and networks, but not in off-target negative controls. Positive associations were observed for degree of the l-DMN and segregation of target networks overall, with significant effects for DMN and VAT.
CONCLUSION: Cognitive improvement following a-iTBS in MCI may be attributable to local and network-level reconfigurations in functional connectivity. These findings will inform larger trials designed to further evaluate the neural mechanisms of a-iTBS for cognition in MCI.
Additional Links: PMID-40252967
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40252967,
year = {2025},
author = {Aghamoosa, S and Nolin, SA and Chen, A and Caulfield, KA and Lopez, J and Rbeiz, K and Fleischmann, HH and Horn, O and Madden, K and Antonucci, M and Revuelta, G and McTeague, LM and Benitez, A},
title = {Accelerated iTBS-Induced Changes in Resting-State Functional Connectivity Correspond with Cognitive Improvement in Amnestic MCI.},
journal = {Brain stimulation},
volume = {},
number = {},
pages = {},
doi = {10.1016/j.brs.2025.04.012},
pmid = {40252967},
issn = {1876-4754},
abstract = {BACKGROUND: Published results of our Phase I safety and feasibility trial of accelerated intermittent theta burst stimulation (a-iTBS) in mild cognitive impairment (MCI) due to Alzheimer's disease showed a large effect-size improvement in cognition.
OBJECTIVE: Further demonstrate target engagement by identifying whether changes in local and network-level functional connectivity relate to the observed cognitive improvement.
METHODS: Eighteen patients with MCI received 3-day a-iTBS (8 sessions/day) to the left dorsolateral prefrontal cortex at Beam F3 (14,400 total pulses) and completed MRI and cognitive testing at pre- and post-treatment. Based on electric field models, we selected 3 stimulated target regions of interest (ROIs) which belonged to the frontoparietal (FPN), default mode (DMN), and ventral attention (VAT) networks (3 target networks). Metrics of resting-state functional connectivity were computed at the ROI level (within-network degree: number of connections) and network level (segregation: strength of connectivity within network relative to other networks). We correlated changes in cognition and connectivity for the target ROIs and networks; off-target ROI (primary visual) and networks serve as negative controls.
RESULTS: Improvements in cognition were associated with connectivity changes in the target ROIs and networks, but not in off-target negative controls. Positive associations were observed for degree of the l-DMN and segregation of target networks overall, with significant effects for DMN and VAT.
CONCLUSION: Cognitive improvement following a-iTBS in MCI may be attributable to local and network-level reconfigurations in functional connectivity. These findings will inform larger trials designed to further evaluate the neural mechanisms of a-iTBS for cognition in MCI.},
}
RevDate: 2025-04-19
Procedure update and interpretation guide for the brain [18]F-FDG PET study.
Revista espanola de medicina nuclear e imagen molecular pii:S2253-8089(25)00048-5 [Epub ahead of print].
The increase in the prevalence of dementia in our clinical setting poses a diagnostic challenge for the Nuclear Medicine specialist. The aim of this study is to review the modifications in the procedure and to describe the usefulness of the use of brain PET with [18]F-FDG in different clinical indications, both classic and emerging ones. The SEMNIM Neuroimaging working group has considered it appropriate to update the procedures available in Spanish, in order to optimize the quality of this technique and the interpretation of its findings, both for the specialist in Nuclear Medicine and the rest of the professionals involved in the diagnosis and treatment of neurodegenerative diseases.
Additional Links: PMID-40252920
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40252920,
year = {2025},
author = {Vigil Díaz, C and Paredes Rodríguez, P and Perissinotti, A and Fernando Guillén, E and López Mora, D and Lojo Ramírez, JA and Aguiar Fernández, P and Camacho Martí, MV and Nieves Cabrera-Martín, M and , },
title = {Procedure update and interpretation guide for the brain [18]F-FDG PET study.},
journal = {Revista espanola de medicina nuclear e imagen molecular},
volume = {},
number = {},
pages = {500159},
doi = {10.1016/j.remnie.2025.500159},
pmid = {40252920},
issn = {2253-8089},
abstract = {The increase in the prevalence of dementia in our clinical setting poses a diagnostic challenge for the Nuclear Medicine specialist. The aim of this study is to review the modifications in the procedure and to describe the usefulness of the use of brain PET with [18]F-FDG in different clinical indications, both classic and emerging ones. The SEMNIM Neuroimaging working group has considered it appropriate to update the procedures available in Spanish, in order to optimize the quality of this technique and the interpretation of its findings, both for the specialist in Nuclear Medicine and the rest of the professionals involved in the diagnosis and treatment of neurodegenerative diseases.},
}
RevDate: 2025-04-19
Reshaping the gut microbiota: A novel oppinion of Eucommiae cortex polysaccharide alleviate learning and memory impairments in Alzheimer's disease.
Journal of advanced research pii:S2090-1232(25)00269-3 [Epub ahead of print].
BACKGROUND: Alzheimer's disease (AD), which is a chronic neurodegenerative disorder, is marked by the progressive deteriorations in learning and memory capabilities. The microbiota-gut-brain axis has come to be regarded as a crucial element in relation to the pathogenesis as well as the treatment of AD. Eucommiae cortex polysaccharides (EPs), being among the most plentiful substances present in the Eucommiae cortex, show the potential to exert immunomodulatory and neuroprotective function. However, whether EPs are protective against AD and their mechanism of action remain to be investigated OBJECTIVES: We hypothesize that EPs can regulate brain glutamine metabolism through gut microbiota and the butyric acid metabolized by them, improve oxidative stress and autophagy in the brain, and thus alleviate AD.
METHODS: In the present study, we used EPs (0.25 % w/w in food) and fecal microbiota transplantation, as well as butyrate supplementation (0.1 M in water), to intervene in AD mice. Multi-omics were used to determine the mechanism by which EPs improve AD-related learning and memory impairments.
RESULTS: Our results suggest that EPs, functioning as a prebiotic, alleviated learning and memory impairments in AD mice. Mechanistically, EPs are able to reshape the gut microbiota, promote the growth of gut microbiota involved in short-chain fatty acid metabolism, particularly butyrate-producing microbes. The butyrate produced by these microbes improves the brain microenvironment by modulating oxidative stress and autophagy mediated by brain glutamate metabolism, improving learning and memory impairments in AD mice, and inhibiting the formation and deposition of beta-amyloid proteins. Fecal microbiota transplantation (FMT) and butyrate supplementation further confirm this conclusion.
CONCLUSIONS: Our results highlighted that EPs can alleviate learning and memory impairments in AD with a gut microbiota-dependent manner and that butyric acid metabolized by butyric acid-metabolizing bacteria in the gut plays a central role in regulating brain glutamine metabolism to improve brain microenvironmental homeostasis. Meanwhile, the present study provides new insights into the treatment of AD with natural products.
Additional Links: PMID-40252828
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40252828,
year = {2025},
author = {Zhao, Y and Zhao, W and Chai, X and Sun, P and Huang, J and Guo, X and Zhang, L and Ren, D and Yi, C and Zhu, X and Zhao, S},
title = {Reshaping the gut microbiota: A novel oppinion of Eucommiae cortex polysaccharide alleviate learning and memory impairments in Alzheimer's disease.},
journal = {Journal of advanced research},
volume = {},
number = {},
pages = {},
doi = {10.1016/j.jare.2025.04.025},
pmid = {40252828},
issn = {2090-1224},
abstract = {BACKGROUND: Alzheimer's disease (AD), which is a chronic neurodegenerative disorder, is marked by the progressive deteriorations in learning and memory capabilities. The microbiota-gut-brain axis has come to be regarded as a crucial element in relation to the pathogenesis as well as the treatment of AD. Eucommiae cortex polysaccharides (EPs), being among the most plentiful substances present in the Eucommiae cortex, show the potential to exert immunomodulatory and neuroprotective function. However, whether EPs are protective against AD and their mechanism of action remain to be investigated OBJECTIVES: We hypothesize that EPs can regulate brain glutamine metabolism through gut microbiota and the butyric acid metabolized by them, improve oxidative stress and autophagy in the brain, and thus alleviate AD.
METHODS: In the present study, we used EPs (0.25 % w/w in food) and fecal microbiota transplantation, as well as butyrate supplementation (0.1 M in water), to intervene in AD mice. Multi-omics were used to determine the mechanism by which EPs improve AD-related learning and memory impairments.
RESULTS: Our results suggest that EPs, functioning as a prebiotic, alleviated learning and memory impairments in AD mice. Mechanistically, EPs are able to reshape the gut microbiota, promote the growth of gut microbiota involved in short-chain fatty acid metabolism, particularly butyrate-producing microbes. The butyrate produced by these microbes improves the brain microenvironment by modulating oxidative stress and autophagy mediated by brain glutamate metabolism, improving learning and memory impairments in AD mice, and inhibiting the formation and deposition of beta-amyloid proteins. Fecal microbiota transplantation (FMT) and butyrate supplementation further confirm this conclusion.
CONCLUSIONS: Our results highlighted that EPs can alleviate learning and memory impairments in AD with a gut microbiota-dependent manner and that butyric acid metabolized by butyric acid-metabolizing bacteria in the gut plays a central role in regulating brain glutamine metabolism to improve brain microenvironmental homeostasis. Meanwhile, the present study provides new insights into the treatment of AD with natural products.},
}
RevDate: 2025-04-19
FABP7 Expression Modulates the Response of Astrocytes to Induced Endotoxemia.
Glia [Epub ahead of print].
Fatty acid binding proteins (FABPs) are a family of small proteins involved in fatty acid (FA) subcellular trafficking. In the adult central nervous system, FABP7, one of the members of this family, is highly expressed in astrocytes and participates in lipid metabolism, regulation of gene expression, and energy homeostasis. Reactive astrocytes in Alzheimer's disease and amyotrophic lateral sclerosis animal models upregulate FABP7 expression. This upregulation may contribute to the pro-inflammatory phenotype that astrocytes display during neurodegeneration and is detrimental for co-cultured neurons. Here, we explore how FABP7 expression modulates astrocyte response to inflammatory stimuli. Our results showed that silencing FABP7 expression in astrocyte cultures before treatment with different inflammatory stimuli decreases the expression of a luciferase reporter expressed under the control of NF-κB -response elements. Correspondingly, FABP7-silenced astrocytes display decreased nuclear translocation of the NF-κB-p65 subunit in response to these stimuli. Moreover, silencing FABP7 decreases the toxicity of stimulated astrocytes toward co-cultured motor neurons. Similar results were obtained after silencing FABP7 in human astrocytes differentiated from induced pluripotent stem cells. Finally, knockdown of astrocytic FABP7 expression in vivo reduces glial activation in the cerebral cortex of mice after systemic bacterial lipopolysaccharide (LPS) administration. In addition, whole transcriptome RNA sequencing analysis from the cerebral cortex of LPS-treated mice showed a differential inflammatory transcriptional profile, with attenuation of NF-κB-dependent transcriptional response after FABP7 knockdown. Together, our results highlight the potential of FABP7 as a target to modulate neuroinflammation in the central nervous system.
Additional Links: PMID-40251832
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40251832,
year = {2025},
author = {Bresque, M and Esteve, D and Balmer, G and Hamilton, HL and Stephany, JS and Pehar, M and Vargas, MR},
title = {FABP7 Expression Modulates the Response of Astrocytes to Induced Endotoxemia.},
journal = {Glia},
volume = {},
number = {},
pages = {},
doi = {10.1002/glia.70023},
pmid = {40251832},
issn = {1098-1136},
support = {R01NS122973/NH/NIH HHS/United States ; R01NS132760/NH/NIH HHS/United States ; },
abstract = {Fatty acid binding proteins (FABPs) are a family of small proteins involved in fatty acid (FA) subcellular trafficking. In the adult central nervous system, FABP7, one of the members of this family, is highly expressed in astrocytes and participates in lipid metabolism, regulation of gene expression, and energy homeostasis. Reactive astrocytes in Alzheimer's disease and amyotrophic lateral sclerosis animal models upregulate FABP7 expression. This upregulation may contribute to the pro-inflammatory phenotype that astrocytes display during neurodegeneration and is detrimental for co-cultured neurons. Here, we explore how FABP7 expression modulates astrocyte response to inflammatory stimuli. Our results showed that silencing FABP7 expression in astrocyte cultures before treatment with different inflammatory stimuli decreases the expression of a luciferase reporter expressed under the control of NF-κB -response elements. Correspondingly, FABP7-silenced astrocytes display decreased nuclear translocation of the NF-κB-p65 subunit in response to these stimuli. Moreover, silencing FABP7 decreases the toxicity of stimulated astrocytes toward co-cultured motor neurons. Similar results were obtained after silencing FABP7 in human astrocytes differentiated from induced pluripotent stem cells. Finally, knockdown of astrocytic FABP7 expression in vivo reduces glial activation in the cerebral cortex of mice after systemic bacterial lipopolysaccharide (LPS) administration. In addition, whole transcriptome RNA sequencing analysis from the cerebral cortex of LPS-treated mice showed a differential inflammatory transcriptional profile, with attenuation of NF-κB-dependent transcriptional response after FABP7 knockdown. Together, our results highlight the potential of FABP7 as a target to modulate neuroinflammation in the central nervous system.},
}
RevDate: 2025-04-18
CmpDate: 2025-04-18
Bacopa monnieri phytochemicals as promising BACE1 inhibitors for Alzheimer's disease therapy.
Scientific reports, 15(1):13504.
Alzheimer's disease (AD) remains a formidable challenge, necessitating the discovery of effective therapeutic agents targeting β-site amyloid precursor protein cleaving enzyme 1 (BACE1). This study investigates the inhibitory potential of phytochemicals derived from Bacopa monnieri, a plant renowned for its cognitive-enhancing properties, in comparison to established synthetic inhibitors such as Atabecestat, Lanabecestat, and Verubecestat. Utilizing molecular docking and advanced computational simulations, we demonstrate that Bacopaside I exhibits superior binding affinity and a unique interaction profile with BACE1, suggesting a more nuanced inhibitory mechanism. Our findings highlight the promising role of Bacopa monnieri phytochemicals as viable alternatives to synthetic drugs, emphasizing their potential to overcome limitations faced in clinical settings. Furthermore, the development of the SIMANA (https://simana.streamlit.app/) platform enhances the visualization and analysis of protein-ligand interactions, facilitating a deeper understanding of the dynamics involved. This research not only underscores the therapeutic promise of natural compounds in AD treatment but also advocates for a paradigm shift towards integrating traditional medicinal knowledge into contemporary drug discovery efforts.
Additional Links: PMID-40251199
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40251199,
year = {2025},
author = {Sangeet, S and Khan, A},
title = {Bacopa monnieri phytochemicals as promising BACE1 inhibitors for Alzheimer's disease therapy.},
journal = {Scientific reports},
volume = {15},
number = {1},
pages = {13504},
pmid = {40251199},
issn = {2045-2322},
mesh = {*Amyloid Precursor Protein Secretases/antagonists & inhibitors/chemistry/metabolism ; *Alzheimer Disease/drug therapy ; *Bacopa/chemistry ; *Aspartic Acid Endopeptidases/antagonists & inhibitors/metabolism/chemistry ; Molecular Docking Simulation ; Humans ; *Phytochemicals/pharmacology/chemistry/therapeutic use ; Protein Binding ; Triterpenes/chemistry/pharmacology ; },
abstract = {Alzheimer's disease (AD) remains a formidable challenge, necessitating the discovery of effective therapeutic agents targeting β-site amyloid precursor protein cleaving enzyme 1 (BACE1). This study investigates the inhibitory potential of phytochemicals derived from Bacopa monnieri, a plant renowned for its cognitive-enhancing properties, in comparison to established synthetic inhibitors such as Atabecestat, Lanabecestat, and Verubecestat. Utilizing molecular docking and advanced computational simulations, we demonstrate that Bacopaside I exhibits superior binding affinity and a unique interaction profile with BACE1, suggesting a more nuanced inhibitory mechanism. Our findings highlight the promising role of Bacopa monnieri phytochemicals as viable alternatives to synthetic drugs, emphasizing their potential to overcome limitations faced in clinical settings. Furthermore, the development of the SIMANA (https://simana.streamlit.app/) platform enhances the visualization and analysis of protein-ligand interactions, facilitating a deeper understanding of the dynamics involved. This research not only underscores the therapeutic promise of natural compounds in AD treatment but also advocates for a paradigm shift towards integrating traditional medicinal knowledge into contemporary drug discovery efforts.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Amyloid Precursor Protein Secretases/antagonists & inhibitors/chemistry/metabolism
*Alzheimer Disease/drug therapy
*Bacopa/chemistry
*Aspartic Acid Endopeptidases/antagonists & inhibitors/metabolism/chemistry
Molecular Docking Simulation
Humans
*Phytochemicals/pharmacology/chemistry/therapeutic use
Protein Binding
Triterpenes/chemistry/pharmacology
RevDate: 2025-04-18
Leveraging mHealth wearables for managing patients with Alzheimer's disease: a scoping review.
Drug discovery today pii:S1359-6446(25)00076-5 [Epub ahead of print].
In this scoping review, we examine the role of wearable devices in diagnosing, treating, and monitoring Alzheimer's disease (AD) and mild cognitive impairment (MCI). It identifies various devices, including fitness trackers, smartwatches, electroencephalographic equipment, and sensors, which are used for monitoring physical activity, sleep patterns, and cognitive functions. Our review highlights the potential of these devices for early diagnosis and treatment, improving patient autonomy and quality of life. However, challenges, such as data privacy, device adherence, and technical limitations, remain. Future research should focus on integrating wearable devices with advanced diagnostic tools and validating their effectiveness across diverse populations.
Additional Links: PMID-40250750
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40250750,
year = {2025},
author = {Noda, K and Kageyama, I and Kobayashi, Y and Lim, Y and Sengoku, S and Kodama, K},
title = {Leveraging mHealth wearables for managing patients with Alzheimer's disease: a scoping review.},
journal = {Drug discovery today},
volume = {},
number = {},
pages = {104363},
doi = {10.1016/j.drudis.2025.104363},
pmid = {40250750},
issn = {1878-5832},
abstract = {In this scoping review, we examine the role of wearable devices in diagnosing, treating, and monitoring Alzheimer's disease (AD) and mild cognitive impairment (MCI). It identifies various devices, including fitness trackers, smartwatches, electroencephalographic equipment, and sensors, which are used for monitoring physical activity, sleep patterns, and cognitive functions. Our review highlights the potential of these devices for early diagnosis and treatment, improving patient autonomy and quality of life. However, challenges, such as data privacy, device adherence, and technical limitations, remain. Future research should focus on integrating wearable devices with advanced diagnostic tools and validating their effectiveness across diverse populations.},
}
RevDate: 2025-04-18
Plumbagin as a potential therapeutic agent for scopolamine-induced Alzheimer's disease: Mechanistic insights into GSK-3β inhibition.
Brain research pii:S0006-8993(25)00209-4 [Epub ahead of print].
BACKGROUND: The study aimed to evaluate Plumbagin's neuroprotective potential against scopolamine-induced Alzheimer's disease, proposing that its effects may involve GSK-3β inhibition, a key factor in tau hyperphosphorylation, to promote neuroprotection in Wistar rats.
METHODS: Alzheimer's was induced in male Wistar rats. After acclimatization, the rats were subjected to daily intraperitoneal treatment with scopolamine (0.7 mg/kg) and oral administration of Plumbagin (10 mg/kg) for 13 days. The cognitive function of treated rats was evaluated using the Morris water maze test, along with assessments of locomotor activity, acetylcholinesterase activity (AChE), protein levels, antioxidant parameters, cytokines and Brain-Derived Neurotrophic Factor (BDNF) and brain histopathology (hippocampus).
RESULTS: The Plumbagin (10 mg/kg, oral) as given orally significantly improved neurobehavioral alterations compared to Alzheimer's induced group. Scopolamine impaired cognitive function and increased locomotor activity ([#]P < 0.05). Treatments improved Morris water maze performance, reducing Escape latency time and increasing Time spent in the target quadrant (*P < 0.05). Biochemically, treatments significantly improved BDNF (*P < 0.05), decreased AChE activity, oxidative stress, reduced Interleukin-6 and Tumor Necrosis Factor Alpha (*P < 0.05) and reversed Scopolamine induced hippocampal neuronal loss ([##]P < 0.01). Plumbagin showed significant (*P < 0.05) neuroprotective effects, improving cognitive function, reducing AChE activity, Malondialdehyde, oxidative stress, and neuroinflammatory markers exceeding individual treatments in the scopolamine-induced Alzheimer's disease model. These improvements suggest a possible mechanism through the inhibition of GSK-3β, which may contribute to the observed neuroprotective effects.
CONCLUSION: This study suggests that Plumbagin's neuroprotective effects in scopolamine-induced Alzheimer's disease may involve GSK-3β inhibition. Plumbagin shows significant therapeutic potential for Alzheimer's treatment, warranting further investigation of its mechanism.
Additional Links: PMID-40250748
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40250748,
year = {2025},
author = {Ahsan, R and Khan, MM and Mishra, A and Noor, G and Ahmad, U},
title = {Plumbagin as a potential therapeutic agent for scopolamine-induced Alzheimer's disease: Mechanistic insights into GSK-3β inhibition.},
journal = {Brain research},
volume = {},
number = {},
pages = {149650},
doi = {10.1016/j.brainres.2025.149650},
pmid = {40250748},
issn = {1872-6240},
abstract = {BACKGROUND: The study aimed to evaluate Plumbagin's neuroprotective potential against scopolamine-induced Alzheimer's disease, proposing that its effects may involve GSK-3β inhibition, a key factor in tau hyperphosphorylation, to promote neuroprotection in Wistar rats.
METHODS: Alzheimer's was induced in male Wistar rats. After acclimatization, the rats were subjected to daily intraperitoneal treatment with scopolamine (0.7 mg/kg) and oral administration of Plumbagin (10 mg/kg) for 13 days. The cognitive function of treated rats was evaluated using the Morris water maze test, along with assessments of locomotor activity, acetylcholinesterase activity (AChE), protein levels, antioxidant parameters, cytokines and Brain-Derived Neurotrophic Factor (BDNF) and brain histopathology (hippocampus).
RESULTS: The Plumbagin (10 mg/kg, oral) as given orally significantly improved neurobehavioral alterations compared to Alzheimer's induced group. Scopolamine impaired cognitive function and increased locomotor activity ([#]P < 0.05). Treatments improved Morris water maze performance, reducing Escape latency time and increasing Time spent in the target quadrant (*P < 0.05). Biochemically, treatments significantly improved BDNF (*P < 0.05), decreased AChE activity, oxidative stress, reduced Interleukin-6 and Tumor Necrosis Factor Alpha (*P < 0.05) and reversed Scopolamine induced hippocampal neuronal loss ([##]P < 0.01). Plumbagin showed significant (*P < 0.05) neuroprotective effects, improving cognitive function, reducing AChE activity, Malondialdehyde, oxidative stress, and neuroinflammatory markers exceeding individual treatments in the scopolamine-induced Alzheimer's disease model. These improvements suggest a possible mechanism through the inhibition of GSK-3β, which may contribute to the observed neuroprotective effects.
CONCLUSION: This study suggests that Plumbagin's neuroprotective effects in scopolamine-induced Alzheimer's disease may involve GSK-3β inhibition. Plumbagin shows significant therapeutic potential for Alzheimer's treatment, warranting further investigation of its mechanism.},
}
RevDate: 2025-04-18
CmpDate: 2025-04-18
Lipid Droplet in Lipodystrophy and Neurodegeneration.
Biology of the cell, 117(4):e70009.
Lipid droplets are ubiquitous yet distinct intracellular organelles that are gaining attention for their uses outside of energy storage. Their formation, role in the physiological function, and the onset of the pathology have been gaining attention recently. Their structure, synthesis, and turnover play dynamic roles in both lipodystrophy and neurodegeneration. Factors like development, aging, inflammation, and cellular stress regulate the synthesis of lipid droplets. The biogenesis of lipid droplets has a critical role in reducing cellular stress. Lipid droplets, in response to stress, sequester hazardous lipids into their neutral lipid core, preserving energy and redox balance while guarding against lipotoxicity. Thus, the maintenance of lipid droplet homeostasis in adipose tissue, CNS, and other body tissues is essential for maintaining organismal health. Insulin resistance, hypertriglyceridemia, and lipid droplet accumulation are the severe metabolic abnormalities that accompany lipodystrophy-related fat deficit. Accumulation of lipid droplets is detected in almost all neurodegenerative diseases like Alzheimer's, Parkinson's, Huntington's, and Hereditary spastic paraplegia. Hence, the regulation of lipid droplets can be used as an alternative approach to the treatment of several diseases. The current review summarizes the structure, composition, biogenesis, and turnover of lipid droplets, with an emphasis on the factors responsible for the accumulation and importance of lipid droplets in lipodystrophy and neurodegenerative disease.
Additional Links: PMID-40249069
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40249069,
year = {2025},
author = {Behera, P and Mishra, M},
title = {Lipid Droplet in Lipodystrophy and Neurodegeneration.},
journal = {Biology of the cell},
volume = {117},
number = {4},
pages = {e70009},
doi = {10.1111/boc.70009},
pmid = {40249069},
issn = {1768-322X},
support = {221610052028//UGC/ ; },
mesh = {Humans ; *Lipodystrophy/metabolism/pathology ; *Lipid Droplets/metabolism/pathology ; *Neurodegenerative Diseases/metabolism/pathology ; Animals ; Lipid Metabolism ; },
abstract = {Lipid droplets are ubiquitous yet distinct intracellular organelles that are gaining attention for their uses outside of energy storage. Their formation, role in the physiological function, and the onset of the pathology have been gaining attention recently. Their structure, synthesis, and turnover play dynamic roles in both lipodystrophy and neurodegeneration. Factors like development, aging, inflammation, and cellular stress regulate the synthesis of lipid droplets. The biogenesis of lipid droplets has a critical role in reducing cellular stress. Lipid droplets, in response to stress, sequester hazardous lipids into their neutral lipid core, preserving energy and redox balance while guarding against lipotoxicity. Thus, the maintenance of lipid droplet homeostasis in adipose tissue, CNS, and other body tissues is essential for maintaining organismal health. Insulin resistance, hypertriglyceridemia, and lipid droplet accumulation are the severe metabolic abnormalities that accompany lipodystrophy-related fat deficit. Accumulation of lipid droplets is detected in almost all neurodegenerative diseases like Alzheimer's, Parkinson's, Huntington's, and Hereditary spastic paraplegia. Hence, the regulation of lipid droplets can be used as an alternative approach to the treatment of several diseases. The current review summarizes the structure, composition, biogenesis, and turnover of lipid droplets, with an emphasis on the factors responsible for the accumulation and importance of lipid droplets in lipodystrophy and neurodegenerative disease.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Lipodystrophy/metabolism/pathology
*Lipid Droplets/metabolism/pathology
*Neurodegenerative Diseases/metabolism/pathology
Animals
Lipid Metabolism
RevDate: 2025-04-18
Repetitive transcranial magnetic stimulation may promote the reversion of mild cognitive impairment to normal cognition.
Frontiers in psychiatry, 16:1544728.
PURPOSE: This study aimed to investigate the potential effects of repetitive transcranial magnetic stimulation (rTMS) on the reversion of mild cognitive impairment (MCI) to normal cognitive function and to elucidate the underlying mechanisms.
METHODS: The study enrolled 25 MCI participants, who underwent a 10-day of rTMS treatment and an 18-month follow-up, along with 15 healthy subjects. Participants with MCI were categorized into MCI reverters (MCI-R) and MCI maintainers (MCI-M). We assessed differences in baseline cognitive performance, functional connectivity, and changes of cognitive functions after rTMS between MCI-R and MCI-M to identify possible predictors of reversion of MCI and explore the neural modulation mechanisms.
RESULTS: MCI-M exhibited more severe cognitive impairments across more domains, particularly in language function (p < 0.05). Functional connectivity was more severely damaged in MCI-M participants, notably within the default mode network (DMN), executive control network (ECN), and frontoparietal network (FPN). After rTMS therapy, MCI-R participants demonstrated more significantly improved immediate and delayed recall memory scores (p < 0.05). These memory function changes and baseline functional connectivity of DMN, ECN, and FPN were predictive of the reversion of MCI.
CONCLUSIONS: The efficacy of rTMS in memory function may promote the reversion of MCI to normal cognition, with the functional connectivity of DMN, ECN, and FPN playing a crucial important role. The severity of cognitive impairment and functional connectivity damage correlated with the likelihood of the reversion of MCI to normal cognition, underscoring the importance of early rTMS intervention for dementia prevention.
Additional Links: PMID-40248597
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40248597,
year = {2025},
author = {Guo, Z and Jiang, Y and He, J and Jiang, N},
title = {Repetitive transcranial magnetic stimulation may promote the reversion of mild cognitive impairment to normal cognition.},
journal = {Frontiers in psychiatry},
volume = {16},
number = {},
pages = {1544728},
pmid = {40248597},
issn = {1664-0640},
abstract = {PURPOSE: This study aimed to investigate the potential effects of repetitive transcranial magnetic stimulation (rTMS) on the reversion of mild cognitive impairment (MCI) to normal cognitive function and to elucidate the underlying mechanisms.
METHODS: The study enrolled 25 MCI participants, who underwent a 10-day of rTMS treatment and an 18-month follow-up, along with 15 healthy subjects. Participants with MCI were categorized into MCI reverters (MCI-R) and MCI maintainers (MCI-M). We assessed differences in baseline cognitive performance, functional connectivity, and changes of cognitive functions after rTMS between MCI-R and MCI-M to identify possible predictors of reversion of MCI and explore the neural modulation mechanisms.
RESULTS: MCI-M exhibited more severe cognitive impairments across more domains, particularly in language function (p < 0.05). Functional connectivity was more severely damaged in MCI-M participants, notably within the default mode network (DMN), executive control network (ECN), and frontoparietal network (FPN). After rTMS therapy, MCI-R participants demonstrated more significantly improved immediate and delayed recall memory scores (p < 0.05). These memory function changes and baseline functional connectivity of DMN, ECN, and FPN were predictive of the reversion of MCI.
CONCLUSIONS: The efficacy of rTMS in memory function may promote the reversion of MCI to normal cognition, with the functional connectivity of DMN, ECN, and FPN playing a crucial important role. The severity of cognitive impairment and functional connectivity damage correlated with the likelihood of the reversion of MCI to normal cognition, underscoring the importance of early rTMS intervention for dementia prevention.},
}
RevDate: 2025-04-18
Efficacy of Home-Based Remotely Supervised Transcranial Direct Current Stimulation for Managing Neuropsychiatric Symptoms in Older Adults With Alzheimer's Disease and Related Dementias.
Integrative and complementary therapies, 30(5):209-219.
BACKGROUND: Neuropsychiatric symptoms (NPS) are prevalent among persons with Alzheimer's disease and related dementias (ADRD). However, there are limited safe and effective nonpharmacological treatments for controlling NPS. Transcranial direct current stimulation (tDCS) is a promising noninvasive and safe treatment.
MATERIALS AND METHODS: This study investigated the effects of remotely supervised tDCS in managing NPS in older adults with mild to moderate ADRD. Forty older adults diagnosed with early-stage ADRD were randomly assigned in a 1:1 ratio to receive home-based active tDCS (n = 20) or sham tDCS (n = 20).
RESULTS: Results showed a significantly greater improvement in the following NPS: scratching (P = 0.052, Hedges' g = -0.60 [confidence interval {CI}: -1.24, 0.04], Cliff's δ = -0.41 [CI: -0.67, -0.06]), nighttime behaviors (P = 0.041; Hedges' g = -0.62 [CI: -1.26, 0.03]; Cliff's δ = -0.41 [CI: -0.67, -0.06], and appetite/eating changes (P = 0.010; Hedges' g = -0.78 [CI: -1.43, -0.13]; Cliff's δ = -0.41 [CI: - 0.56, -0.10]).
CONCLUSION: This study shows promising initial results for using home-based, remotely supervised tDCS to manage NPS, such as nighttime behaviors, changes in eating and appetite, and scratching. Larger studies with more participants are needed to explore various tDCS doses and their long-term effects on NPS.
Additional Links: PMID-40248168
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40248168,
year = {2024},
author = {Park, J and Lee, C and Lin, L and Galvin, J and Fain, MJ and Allen, A and Park, L and Ahn, H},
title = {Efficacy of Home-Based Remotely Supervised Transcranial Direct Current Stimulation for Managing Neuropsychiatric Symptoms in Older Adults With Alzheimer's Disease and Related Dementias.},
journal = {Integrative and complementary therapies},
volume = {30},
number = {5},
pages = {209-219},
pmid = {40248168},
issn = {2768-3206},
abstract = {BACKGROUND: Neuropsychiatric symptoms (NPS) are prevalent among persons with Alzheimer's disease and related dementias (ADRD). However, there are limited safe and effective nonpharmacological treatments for controlling NPS. Transcranial direct current stimulation (tDCS) is a promising noninvasive and safe treatment.
MATERIALS AND METHODS: This study investigated the effects of remotely supervised tDCS in managing NPS in older adults with mild to moderate ADRD. Forty older adults diagnosed with early-stage ADRD were randomly assigned in a 1:1 ratio to receive home-based active tDCS (n = 20) or sham tDCS (n = 20).
RESULTS: Results showed a significantly greater improvement in the following NPS: scratching (P = 0.052, Hedges' g = -0.60 [confidence interval {CI}:
-1.24, 0.04], Cliff's δ = -0.41 [CI: -0.67, -0.06]), nighttime behaviors (P = 0.041; Hedges' g = -0.62 [CI: -1.26, 0.03]; Cliff's δ = -0.41 [CI: -0.67, -0.06], and appetite/eating changes (P = 0.010; Hedges' g = -0.78 [CI: -1.43, -0.13]; Cliff's δ = -0.41 [CI: - 0.56, -0.10]).
CONCLUSION: This study shows promising initial results for using home-based, remotely supervised tDCS to manage NPS, such as nighttime behaviors, changes in eating and appetite, and scratching. Larger studies with more participants are needed to explore various tDCS doses and their long-term effects on NPS.},
}
RevDate: 2025-04-18
A Deep Subgrouping Framework for Precision Drug Repurposing via Emulating Clinical Trials on Real-world Patient Data.
KDD : proceedings. International Conference on Knowledge Discovery & Data Mining, 2025(v1):2347-2358.
Drug repurposing identifies new therapeutic uses for existing drugs, reducing the time and costs compared to traditional de novo drug discovery. Most existing drug repurposing studies using real-world patient data often treat the entire population as homogeneous, ignoring the heterogeneity of treatment responses across patient subgroups. This approach may overlook promising drugs that benefit specific subgroups but lack notable treatment effects across the entire population, potentially limiting the number of repurposable candidates identified. To address this, we introduce STEDR, a novel drug repurposing framework that integrates subgroup analysis with treatment effect estimation. Our approach first identifies repurposing candidates by emulating multiple clinical trials on real-world patient data and then characterizes patient subgroups by learning subgroup-specific treatment effects. We deploy STEDR to Alzheimer's Disease (AD), a condition with few approved drugs and known heterogeneity in treatment responses. We emulate trials for over one thousand medications on a large-scale real-world database covering over 8 million patients, identifying 14 drug candidates with beneficial effects to AD in characterized subgroups. Experiments demonstrate STEDR's superior capability in identifying repurposing candidates compared to existing approaches. Additionally, our method can characterize clinically relevant patient subgroups associated with important AD-related risk factors, paving the way for precision drug repurposing.
Additional Links: PMID-40248108
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40248108,
year = {2025},
author = {Lee, S and Liu, R and Cheng, F and Zhang, P},
title = {A Deep Subgrouping Framework for Precision Drug Repurposing via Emulating Clinical Trials on Real-world Patient Data.},
journal = {KDD : proceedings. International Conference on Knowledge Discovery & Data Mining},
volume = {2025},
number = {v1},
pages = {2347-2358},
pmid = {40248108},
issn = {2154-817X},
abstract = {Drug repurposing identifies new therapeutic uses for existing drugs, reducing the time and costs compared to traditional de novo drug discovery. Most existing drug repurposing studies using real-world patient data often treat the entire population as homogeneous, ignoring the heterogeneity of treatment responses across patient subgroups. This approach may overlook promising drugs that benefit specific subgroups but lack notable treatment effects across the entire population, potentially limiting the number of repurposable candidates identified. To address this, we introduce STEDR, a novel drug repurposing framework that integrates subgroup analysis with treatment effect estimation. Our approach first identifies repurposing candidates by emulating multiple clinical trials on real-world patient data and then characterizes patient subgroups by learning subgroup-specific treatment effects. We deploy STEDR to Alzheimer's Disease (AD), a condition with few approved drugs and known heterogeneity in treatment responses. We emulate trials for over one thousand medications on a large-scale real-world database covering over 8 million patients, identifying 14 drug candidates with beneficial effects to AD in characterized subgroups. Experiments demonstrate STEDR's superior capability in identifying repurposing candidates compared to existing approaches. Additionally, our method can characterize clinically relevant patient subgroups associated with important AD-related risk factors, paving the way for precision drug repurposing.},
}
RevDate: 2025-04-18
Engineered nanoparticles for the treatment of Alzheimer's disease.
Frontiers in pharmacology, 16:1510798.
Alzheimer's disease (AD) is one of the most common diseases characterized by neurodegeneration and is becoming a major public health problem worldwide. AD is manifested mainly by progressive impairments in cognition, emotion, language and memory in the elderly population. Many treatment strategies have been explored for decades; however, there is still no effective way to address the root cause of AD pathogenesis, only to target symptoms to improve patient cognitive outcomes. Intracerebral administration is difficult because of the challenges posed by the blood‒brain barrier (BBB). NPs are materials with sizes between 1 and 100 nm that can improve biocompatibility, extend the half-life, transport macromolecules, be delivered across the BBB to the central nervous system, and exhibit good targeting capabilities. NPs can provide new ideas for the treatment of AD in terms of their antiaging, antineuroinflammatory, antioxidative, and nerve repair-promoting effects. In this manuscript, we first describe the relationship between AD and the BBB. Second, we introduce the application of nanoparticles for AD treatment. Finally, we summarize the challenges faced by nanoparticles in the treatment of AD.
Additional Links: PMID-40248097
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40248097,
year = {2025},
author = {Jia, J and Zhao, S and Zhao, J and Gao, Y},
title = {Engineered nanoparticles for the treatment of Alzheimer's disease.},
journal = {Frontiers in pharmacology},
volume = {16},
number = {},
pages = {1510798},
pmid = {40248097},
issn = {1663-9812},
abstract = {Alzheimer's disease (AD) is one of the most common diseases characterized by neurodegeneration and is becoming a major public health problem worldwide. AD is manifested mainly by progressive impairments in cognition, emotion, language and memory in the elderly population. Many treatment strategies have been explored for decades; however, there is still no effective way to address the root cause of AD pathogenesis, only to target symptoms to improve patient cognitive outcomes. Intracerebral administration is difficult because of the challenges posed by the blood‒brain barrier (BBB). NPs are materials with sizes between 1 and 100 nm that can improve biocompatibility, extend the half-life, transport macromolecules, be delivered across the BBB to the central nervous system, and exhibit good targeting capabilities. NPs can provide new ideas for the treatment of AD in terms of their antiaging, antineuroinflammatory, antioxidative, and nerve repair-promoting effects. In this manuscript, we first describe the relationship between AD and the BBB. Second, we introduce the application of nanoparticles for AD treatment. Finally, we summarize the challenges faced by nanoparticles in the treatment of AD.},
}
RevDate: 2025-04-18
Biofilm Biology to Brain Health: Harnessing Microbial Wisdom to Uncover Amyloid Dissociating Bifunctional Nano Chaperones for Alzheimer's Therapeutics.
ACS chemical neuroscience [Epub ahead of print].
Microbial infections have long been implicated in the gut-brain link to Alzheimer's disease (AD). These infections may influence AD development either directly, through brain invasion, or indirectly via bacterial metabolites crossing the blood-brain-barrier (BBB) or interacting with the enteric nervous system (ENS). Such findings have inspired clinicians to repurpose antimicrobial drugs for AD, yielding promising results. However, the sole bacterial link to AD may be insufficiently understood. Bacterial amyloid presence in biofilms is well-documented, with certain bacterial proteins exacerbating amyloid formation while others inhibit it. For instance, Curli-specific gene protein C (CsgC) in E. coli suppresses curli amyloid formation. This review investigates the possibility of human CsgC-like proteins, identifying beta-2 microglobulin (β2M) and E3 ubiquitin ligases (E3s) as potential analogs that may influence amyloid degradation. We propose that nanoparticles (NPs) could serve as platforms to anchor these proteins, forming Amyloid Dissociating Bifunctional NanoChaperones (ADBiNaCs) with enhanced antiamyloidogenic activity. This innovative approach holds promise for novel AD treatment strategies, meriting further investigation into the role of bacterial and human amyloid-modulating proteins in AD pathology.
Additional Links: PMID-40247766
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40247766,
year = {2025},
author = {Randhawa, S and Saini, TC and Bathla, M and Teji, N and Acharya, A},
title = {Biofilm Biology to Brain Health: Harnessing Microbial Wisdom to Uncover Amyloid Dissociating Bifunctional Nano Chaperones for Alzheimer's Therapeutics.},
journal = {ACS chemical neuroscience},
volume = {},
number = {},
pages = {},
doi = {10.1021/acschemneuro.4c00868},
pmid = {40247766},
issn = {1948-7193},
abstract = {Microbial infections have long been implicated in the gut-brain link to Alzheimer's disease (AD). These infections may influence AD development either directly, through brain invasion, or indirectly via bacterial metabolites crossing the blood-brain-barrier (BBB) or interacting with the enteric nervous system (ENS). Such findings have inspired clinicians to repurpose antimicrobial drugs for AD, yielding promising results. However, the sole bacterial link to AD may be insufficiently understood. Bacterial amyloid presence in biofilms is well-documented, with certain bacterial proteins exacerbating amyloid formation while others inhibit it. For instance, Curli-specific gene protein C (CsgC) in E. coli suppresses curli amyloid formation. This review investigates the possibility of human CsgC-like proteins, identifying beta-2 microglobulin (β2M) and E3 ubiquitin ligases (E3s) as potential analogs that may influence amyloid degradation. We propose that nanoparticles (NPs) could serve as platforms to anchor these proteins, forming Amyloid Dissociating Bifunctional NanoChaperones (ADBiNaCs) with enhanced antiamyloidogenic activity. This innovative approach holds promise for novel AD treatment strategies, meriting further investigation into the role of bacterial and human amyloid-modulating proteins in AD pathology.},
}
RevDate: 2025-04-18
CmpDate: 2025-04-18
Sex differences in brain iron deposition and microglial ferritin in Alzheimer's disease.
Science progress, 108(2):368504251336080.
ObjectiveIron is the most abundant metal in the human brain, and plays a crucial role in many biological processes. However, disruptions in brain iron metabolism can lead to iron buildup, which occurs with aging and is linked to several brain disorders, including Alzheimer's disease. Microglia, the brain's resident immune cells, have the highest capacity to store iron, which is stored intracellularly within ferritin complexes. Importantly, women are at a higher risk of developing Alzheimer's disease and experience faster disease progression compared to men.MethodsWe used postmortem brain samples from patients with Alzheimer's disease and small vessel disease patients of both sexes for immunohistochemical studies. Samples were stained with the Prussian blue method to visualize iron deposits and with antibodies against the microglia marker Iba1 and ferritin light chain.ResultsOur study reveals that the number of iron deposits and the levels of ferritin light chain in microglia are positively correlated in men with Alzheimer's disease, but negatively correlated in women. There is no correlation between brain iron deposition and ferritin in samples from patients with small vessel disease of both sexes.ConclusionsThese results could inform more tailored approaches to the treatment and management of Alzheimer's disease based on sex-specific differences in brain iron metabolism and microglial iron storage capacity.
Additional Links: PMID-40247604
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40247604,
year = {2025},
author = {Rahman, SM and Tan, C and Kakita, A and Moruno-Manchon, JF},
title = {Sex differences in brain iron deposition and microglial ferritin in Alzheimer's disease.},
journal = {Science progress},
volume = {108},
number = {2},
pages = {368504251336080},
doi = {10.1177/00368504251336080},
pmid = {40247604},
issn = {2047-7163},
mesh = {Humans ; *Alzheimer Disease/metabolism/pathology ; *Microglia/metabolism/pathology ; Female ; Male ; *Iron/metabolism ; *Brain/metabolism/pathology ; Aged ; *Ferritins/metabolism ; *Sex Characteristics ; Aged, 80 and over ; Apoferritins/metabolism ; Sex Factors ; },
abstract = {ObjectiveIron is the most abundant metal in the human brain, and plays a crucial role in many biological processes. However, disruptions in brain iron metabolism can lead to iron buildup, which occurs with aging and is linked to several brain disorders, including Alzheimer's disease. Microglia, the brain's resident immune cells, have the highest capacity to store iron, which is stored intracellularly within ferritin complexes. Importantly, women are at a higher risk of developing Alzheimer's disease and experience faster disease progression compared to men.MethodsWe used postmortem brain samples from patients with Alzheimer's disease and small vessel disease patients of both sexes for immunohistochemical studies. Samples were stained with the Prussian blue method to visualize iron deposits and with antibodies against the microglia marker Iba1 and ferritin light chain.ResultsOur study reveals that the number of iron deposits and the levels of ferritin light chain in microglia are positively correlated in men with Alzheimer's disease, but negatively correlated in women. There is no correlation between brain iron deposition and ferritin in samples from patients with small vessel disease of both sexes.ConclusionsThese results could inform more tailored approaches to the treatment and management of Alzheimer's disease based on sex-specific differences in brain iron metabolism and microglial iron storage capacity.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alzheimer Disease/metabolism/pathology
*Microglia/metabolism/pathology
Female
Male
*Iron/metabolism
*Brain/metabolism/pathology
Aged
*Ferritins/metabolism
*Sex Characteristics
Aged, 80 and over
Apoferritins/metabolism
Sex Factors
RevDate: 2025-04-17
Extracellular vesicle-based point-of-care testing for diagnosis and monitoring of Alzheimer's disease.
Microsystems & nanoengineering, 11(1):65.
Extracellular vesicles (EVs) show potential for early diagnosis of Alzheimer's disease (AD) and monitoring of its progression. However, EV-based AD diagnosis faces challenges due to the small size and low abundance of biomarkers. Here, we report a fully integrated organic electrochemical transistor (OECT) sensor for ultrafast, accurate, and convenient point-of-care testing (POCT) of serum EVs from AD patients. By utilizing acoustoelectric enrichment, the EVs can be quickly propelled, significantly enriched, and specifically bound to the OECT detection area, achieving a gain of over 280 times response in 30 s. The integrated POCT sensor can detect serum EVs from AD patients with a limit of detection as low as 500 EV particles/mL and a reduced detection time of just two minutes. Furthermore, the integrated POCT sensors were used to monitor AD progression in an AD mouse model by testing the mouse Aβ EVs at different time courses (up to 18 months) and compared with the Aβ accumulation using high-resolution magnetic resonance imaging (MRI). This innovative technology has the potential for accurate and rapid diagnosis of Alzheimer's and other neurodegenerative diseases, and monitoring of disease progression and treatment response.
Additional Links: PMID-40246821
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40246821,
year = {2025},
author = {Li, X and Chen, J and Yang, Y and Cai, H and Ao, Z and Xing, Y and Li, K and Yang, K and Guan, W and Friend, J and Lee, LP and Wang, N and Guo, F},
title = {Extracellular vesicle-based point-of-care testing for diagnosis and monitoring of Alzheimer's disease.},
journal = {Microsystems & nanoengineering},
volume = {11},
number = {1},
pages = {65},
pmid = {40246821},
issn = {2055-7434},
abstract = {Extracellular vesicles (EVs) show potential for early diagnosis of Alzheimer's disease (AD) and monitoring of its progression. However, EV-based AD diagnosis faces challenges due to the small size and low abundance of biomarkers. Here, we report a fully integrated organic electrochemical transistor (OECT) sensor for ultrafast, accurate, and convenient point-of-care testing (POCT) of serum EVs from AD patients. By utilizing acoustoelectric enrichment, the EVs can be quickly propelled, significantly enriched, and specifically bound to the OECT detection area, achieving a gain of over 280 times response in 30 s. The integrated POCT sensor can detect serum EVs from AD patients with a limit of detection as low as 500 EV particles/mL and a reduced detection time of just two minutes. Furthermore, the integrated POCT sensors were used to monitor AD progression in an AD mouse model by testing the mouse Aβ EVs at different time courses (up to 18 months) and compared with the Aβ accumulation using high-resolution magnetic resonance imaging (MRI). This innovative technology has the potential for accurate and rapid diagnosis of Alzheimer's and other neurodegenerative diseases, and monitoring of disease progression and treatment response.},
}
RevDate: 2025-04-17
Using machine learning and electronic health record (EHR) data for the early prediction of Alzheimer's Disease and Related Dementias.
The journal of prevention of Alzheimer's disease pii:S2274-5807(25)00114-1 [Epub ahead of print].
BACKGROUND: Over 6 million patients in the United States are affected by Alzheimer's Disease and Related Dementias (ADRD). Early detection of ADRD can significantly improve patient outcomes through timely treatment.
OBJECTIVE: To develop and validate machine learning (ML) models for early ADRD diagnosis and prediction using de-identified EHR data from the University of Missouri (MU) Healthcare.
DESIGN: Retrospective case-control study.
SETTING: The study used de-identified EHR data provided by the MU NextGen Biomedical Informatics, modeled with the PCORnet Common Data Model (CDM).
PARTICIPANTS: An initial cohort of 380,269 patients aged 40 or older with at least two healthcare encounters was narrowed to a final dataset of 4,012 ADRD cases and 119,723 controls.
METHODS: Six ML classifier models: Gradient-Boosted Trees (GBT), Light Gradient-Boosting Machine (LightGBM), Random Forest (RF), eXtreme Gradient-Boosting (XGBoost), Logistic Regression (LR), and Adaptive Boosting (AdaBoost) were evaluated using Area Under the Receiver Operating Characteristic Curve (AUC-ROC), accuracy, sensitivity, specificity, and F1 score. SHAP (SHapley Additive exPlanations) analysis was applied to interpret predictions.
RESULTS: The GBT model achieved the best AUC-ROC scores of 0.809-0.833 across 1- to 5-year prediction windows. SHAP analysis identified depressive disorder, age groups 80-90 yrs and 70-80 yrs, heart disease, anxiety, and the novel risk factors of sleep apnea, and headache.
CONCLUSION: This study underscores the potential of ML models for leveraging EHR data to enable early ADRD prediction, supporting timely interventions, and improving patient outcomes. By identifying both established and novel risk factors, these findings offer new opportunities for personalized screening and management strategies, advancing both clinical and informatics science.
Additional Links: PMID-40246680
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40246680,
year = {2025},
author = {Akter, S and Liu, Z and Simoes, EJ and Rao, P},
title = {Using machine learning and electronic health record (EHR) data for the early prediction of Alzheimer's Disease and Related Dementias.},
journal = {The journal of prevention of Alzheimer's disease},
volume = {},
number = {},
pages = {100169},
doi = {10.1016/j.tjpad.2025.100169},
pmid = {40246680},
issn = {2426-0266},
abstract = {BACKGROUND: Over 6 million patients in the United States are affected by Alzheimer's Disease and Related Dementias (ADRD). Early detection of ADRD can significantly improve patient outcomes through timely treatment.
OBJECTIVE: To develop and validate machine learning (ML) models for early ADRD diagnosis and prediction using de-identified EHR data from the University of Missouri (MU) Healthcare.
DESIGN: Retrospective case-control study.
SETTING: The study used de-identified EHR data provided by the MU NextGen Biomedical Informatics, modeled with the PCORnet Common Data Model (CDM).
PARTICIPANTS: An initial cohort of 380,269 patients aged 40 or older with at least two healthcare encounters was narrowed to a final dataset of 4,012 ADRD cases and 119,723 controls.
METHODS: Six ML classifier models: Gradient-Boosted Trees (GBT), Light Gradient-Boosting Machine (LightGBM), Random Forest (RF), eXtreme Gradient-Boosting (XGBoost), Logistic Regression (LR), and Adaptive Boosting (AdaBoost) were evaluated using Area Under the Receiver Operating Characteristic Curve (AUC-ROC), accuracy, sensitivity, specificity, and F1 score. SHAP (SHapley Additive exPlanations) analysis was applied to interpret predictions.
RESULTS: The GBT model achieved the best AUC-ROC scores of 0.809-0.833 across 1- to 5-year prediction windows. SHAP analysis identified depressive disorder, age groups 80-90 yrs and 70-80 yrs, heart disease, anxiety, and the novel risk factors of sleep apnea, and headache.
CONCLUSION: This study underscores the potential of ML models for leveraging EHR data to enable early ADRD prediction, supporting timely interventions, and improving patient outcomes. By identifying both established and novel risk factors, these findings offer new opportunities for personalized screening and management strategies, advancing both clinical and informatics science.},
}
RevDate: 2025-04-18
Rationale and Design of STAMINA: Senolytics To Alleviate Mobility Issues and Neurological Impairments in Aging, A Geroscience Feasibility Study.
Translational medicine of aging, 7:109-117.
The process of cellular senescence is hypothesized to play a critical role in the development of age-related mobility and cognitive impairments, both of which precede the development of Alzheimer's disease. Therefore, senolytic compounds that eliminate senescent cells represent an alternative strategy that may help improve mobility and cognition in older adults; however, clinical trials are lacking. The goal of this paper is to describe the rationale and study design of a 12-week single arm, open label, pre-post pilot study that administers intermittent doses of two senolytic compounds, Dasatinib and quercetin (DQ), in 12 older adults ≥ 65 years with slow gait speed (<1.0 m/sec) and mild cognitive impairment. Eligible participants are asked to take 1250 mg of and 100 mg of Dasatinib orally once a day for 2 days every 2 weeks, for 6 cycles over 12 consecutive weeks. Both physical and cognitive functional assessments are administered before treatment, as well as 6- and 12- weeks after treatment. Blood and urine samples are taken pre- and post-treatment to assess biomarkers of cellular senescence. The primary outcomes of this trial are feasibility and safety of the intervention, as well as preliminary efficacy on several clinical outcomes (e.g., cerebral blood flow velocity, gait speed, and biomarkers of cellular senescence). The study is approved by the Advarra IRB (#Pro00053594) and a Data Safety Monitoring Board. It is registered at Clinicaltrials.gov (Identifier:NCT05422885). The future results of this study may identify a novel approach for improving mobility and cognition in older adults, thereby preventing progression to Alzheimer's disease.
Additional Links: PMID-40248131
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40248131,
year = {2023},
author = {Millar, CL and Iloputaife, I and Baldyga, K and Kuo, J and Tchkonia, T and Kirkland, JL and Travison, TG and Lipsitz, LA},
title = {Rationale and Design of STAMINA: Senolytics To Alleviate Mobility Issues and Neurological Impairments in Aging, A Geroscience Feasibility Study.},
journal = {Translational medicine of aging},
volume = {7},
number = {},
pages = {109-117},
pmid = {40248131},
issn = {2468-5011},
abstract = {The process of cellular senescence is hypothesized to play a critical role in the development of age-related mobility and cognitive impairments, both of which precede the development of Alzheimer's disease. Therefore, senolytic compounds that eliminate senescent cells represent an alternative strategy that may help improve mobility and cognition in older adults; however, clinical trials are lacking. The goal of this paper is to describe the rationale and study design of a 12-week single arm, open label, pre-post pilot study that administers intermittent doses of two senolytic compounds, Dasatinib and quercetin (DQ), in 12 older adults ≥ 65 years with slow gait speed (<1.0 m/sec) and mild cognitive impairment. Eligible participants are asked to take 1250 mg of and 100 mg of Dasatinib orally once a day for 2 days every 2 weeks, for 6 cycles over 12 consecutive weeks. Both physical and cognitive functional assessments are administered before treatment, as well as 6- and 12- weeks after treatment. Blood and urine samples are taken pre- and post-treatment to assess biomarkers of cellular senescence. The primary outcomes of this trial are feasibility and safety of the intervention, as well as preliminary efficacy on several clinical outcomes (e.g., cerebral blood flow velocity, gait speed, and biomarkers of cellular senescence). The study is approved by the Advarra IRB (#Pro00053594) and a Data Safety Monitoring Board. It is registered at Clinicaltrials.gov (Identifier:NCT05422885). The future results of this study may identify a novel approach for improving mobility and cognition in older adults, thereby preventing progression to Alzheimer's disease.},
}
RevDate: 2025-04-17
Guanylhydrazone and semicarbazone derivatives as potential prototypes for the design of cholinesterase inhibitors against Alzheimer's disease: biological evaluation and molecular modeling studies.
Chemico-biological interactions pii:S0009-2797(25)00145-0 [Epub ahead of print].
Despite being present in many drugs, guanylhydrazones and semicarbazones are two functional groups that have been little investigated as potential therapeutic strategies for the treatment of Alzheimer's disease (AD). For this reason, we initiated the synthesis and evaluation of these compounds as potential anticholinesterase agents, aiming to offer new alternatives for drug development against AD. In the severe phase of AD, butyrylcholinesterase (BChE) becomes the main enzyme responsible for the hydrolysis of acetylcholine (ACh). Therefore, in this project, we present the results of BChE inhibitory activity, enzyme kinetics, cytotoxicity, and molecular modeling studies for three guanylhydrazone and two semicarbazone derivatives that were previously synthesized and evaluated as acetylcholinesterase (AChE) inhibitors. Among the compounds tested, guanylhydrazones (1, 2, and 3) showed inhibitory activity against BChE, exhibiting a mixed non-competitive inhibition profile. Specifically, compound 2 (phenanthrenequinone) demonstrated superior inhibitory potency with an IC50 of 0.68 μM, compared to compound 1 (acridinone) with an IC50 of 3.87 μM, and compound 3 (benzodioxole) with an IC50 of 101.7 μM. In contrast, semicarbazones (4 and 5) showed no BChE inhibition up to the highest concentration tested (300 μM). Importantly, all five compounds were found to be non-cytotoxic. Our results suggest that these compounds have potential as drug prototypes targeting different phases of AD. Compounds 3, 4, and 5 may be more effective in the early phase, when AChE activity remains high; compound 1 could be useful in the intermediate phase; and compound 2 appears particularly promising for the severe phase, when BChE plays a more dominant role.
Additional Links: PMID-40246050
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40246050,
year = {2025},
author = {Neto, DCF and de Almeida, JSFD and Guimarães, SJA and Dos Santos, EM and Nascimento, MDDSB and de Azevedo-Santos, APS and França, TCC and LaPlante, SR and do Nascimento, CJ and Lima, JA},
title = {Guanylhydrazone and semicarbazone derivatives as potential prototypes for the design of cholinesterase inhibitors against Alzheimer's disease: biological evaluation and molecular modeling studies.},
journal = {Chemico-biological interactions},
volume = {},
number = {},
pages = {111515},
doi = {10.1016/j.cbi.2025.111515},
pmid = {40246050},
issn = {1872-7786},
abstract = {Despite being present in many drugs, guanylhydrazones and semicarbazones are two functional groups that have been little investigated as potential therapeutic strategies for the treatment of Alzheimer's disease (AD). For this reason, we initiated the synthesis and evaluation of these compounds as potential anticholinesterase agents, aiming to offer new alternatives for drug development against AD. In the severe phase of AD, butyrylcholinesterase (BChE) becomes the main enzyme responsible for the hydrolysis of acetylcholine (ACh). Therefore, in this project, we present the results of BChE inhibitory activity, enzyme kinetics, cytotoxicity, and molecular modeling studies for three guanylhydrazone and two semicarbazone derivatives that were previously synthesized and evaluated as acetylcholinesterase (AChE) inhibitors. Among the compounds tested, guanylhydrazones (1, 2, and 3) showed inhibitory activity against BChE, exhibiting a mixed non-competitive inhibition profile. Specifically, compound 2 (phenanthrenequinone) demonstrated superior inhibitory potency with an IC50 of 0.68 μM, compared to compound 1 (acridinone) with an IC50 of 3.87 μM, and compound 3 (benzodioxole) with an IC50 of 101.7 μM. In contrast, semicarbazones (4 and 5) showed no BChE inhibition up to the highest concentration tested (300 μM). Importantly, all five compounds were found to be non-cytotoxic. Our results suggest that these compounds have potential as drug prototypes targeting different phases of AD. Compounds 3, 4, and 5 may be more effective in the early phase, when AChE activity remains high; compound 1 could be useful in the intermediate phase; and compound 2 appears particularly promising for the severe phase, when BChE plays a more dominant role.},
}
RevDate: 2025-04-17
The prevalence and influencing factors of reversion from mild cognitive impairment to normal cognition: A systemic review and meta-analysis.
Geriatric nursing (New York, N.Y.), 63:379-387 pii:S0197-4572(25)00104-1 [Epub ahead of print].
OBJECTIVE: The aim was to investigate the pooled prevalence of reversion from mild cognitive impairment (MCI) to normal cognition people, and the influencing factors for reversion.
METHODS: PubMed, Embase, Web of Science, The Cochrane Library, Wanfang Database, China Knowledge Resource Integrated Database (CNKI), Weipu Database, SinoMed were systematically searched from the inception to June 1, 2023. Participants were diagnosis as MCI adults.
RESULTS: In total, 4075 studies were screened and data from 48 studies involving 31876 subjects were used in meta-analysis. The pooled prevalence of reversion from MCI to normal cognition was 31%. The following risk factors were associated with the reversion from MCI to normal cognition: education (low to high), age, Mini-Mental State Examination (MMSE), Functional Activities Questionnaire (FAQ), Auditory Verbal Learning Test (AVLT) delay recall test, Apolipoprotein E (APOE) positive, multiple domain impaired, live along, depression, doing house work daily/exercise once a week.
CONCLUSIONS: The study shows the pooled prevalence of reversion from MCI to normal cognition was high, and there are controllable factors. Understanding the controllable factors of reversion from MCI to normal cognition can provide the clinicians with the theoretical basis for the management and treatment of the patients.
Additional Links: PMID-40245823
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40245823,
year = {2025},
author = {Zhao, Y and Wei, S and Liu, Y and He, X and Li, J and Gao, T and Wang, X and Li, Y and Nan, J and Wang, Y and Ma, Y},
title = {The prevalence and influencing factors of reversion from mild cognitive impairment to normal cognition: A systemic review and meta-analysis.},
journal = {Geriatric nursing (New York, N.Y.)},
volume = {63},
number = {},
pages = {379-387},
doi = {10.1016/j.gerinurse.2025.03.013},
pmid = {40245823},
issn = {1528-3984},
abstract = {OBJECTIVE: The aim was to investigate the pooled prevalence of reversion from mild cognitive impairment (MCI) to normal cognition people, and the influencing factors for reversion.
METHODS: PubMed, Embase, Web of Science, The Cochrane Library, Wanfang Database, China Knowledge Resource Integrated Database (CNKI), Weipu Database, SinoMed were systematically searched from the inception to June 1, 2023. Participants were diagnosis as MCI adults.
RESULTS: In total, 4075 studies were screened and data from 48 studies involving 31876 subjects were used in meta-analysis. The pooled prevalence of reversion from MCI to normal cognition was 31%. The following risk factors were associated with the reversion from MCI to normal cognition: education (low to high), age, Mini-Mental State Examination (MMSE), Functional Activities Questionnaire (FAQ), Auditory Verbal Learning Test (AVLT) delay recall test, Apolipoprotein E (APOE) positive, multiple domain impaired, live along, depression, doing house work daily/exercise once a week.
CONCLUSIONS: The study shows the pooled prevalence of reversion from MCI to normal cognition was high, and there are controllable factors. Understanding the controllable factors of reversion from MCI to normal cognition can provide the clinicians with the theoretical basis for the management and treatment of the patients.},
}
RevDate: 2025-04-17
Antipsychotic-like pharmacological profile of the low impact ampakine CX691 (farampator): Implications for the use of low impact ampakines in the treatment of schizophrenia.
Journal of psychiatric research, 186:145-153 pii:S0022-3956(25)00243-2 [Epub ahead of print].
Ampakines, positive allosteric modulators of AMPA-glutamate receptors (AMPAR), have therapeutic implications in neuropsychiatric and neurological disorders in which AMPAR signaling is compromised such as Alzheimer's, ADHD, and schizophrenia. Low impact ampakines are a distinct subset of ampakines that only partially offset receptor desensitization and do not meaningfully alter binding affinity of AMPAR agonists, which may explain their lack of seizurogenic effects seen with other AMPAR positive modulators. Herein, we describe the preclinical pharmacology and antipsychotic activity of the low impact ampakine 1-(benzofurazan-5-ylcarbonyl)piperidine (CX691). CX691 moderately offsets desensitization in hippocampal patches, supporting its designation as a low impact ampakine and penetrates the blood-brain barrier. CX691 is more potent than well characterized high impact ampakines CX614 and CX546 and low impact ampakine CX516 in abrogating amphetamine-stimulated locomotor activity in Sprague Dawley rats. Low-dose CX691 synergistically reduces methamphetamine-induced locomotor activity when paired with approved antipsychotics clozapine and olanzapine. In rats treated chronically with amphetamine, CX691 retains antipsychotic activity whereas high doses of CX516 lack therapeutic effects. In contrast to haloperidol, CX691 is devoid of cataleptic activity at supratherapeutic doses in rats. CX691 enhances performance in the eight-arm radial maze, a spatial task that assesses hippocampal function, an activity of potential value for ameliorating cognitive deficits in schizophrenia. Taken together, these findings illustrate that low impact ampakines with improved potency might be useful therapeutic interventions in schizophrenic patients when given alone or as adjuncts to ongoing traditional antipsychotic drug therapies.
Additional Links: PMID-40245529
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40245529,
year = {2025},
author = {Radin, DP and Cerne, R and Smith, JL and Witkin, JM and Lippa, A},
title = {Antipsychotic-like pharmacological profile of the low impact ampakine CX691 (farampator): Implications for the use of low impact ampakines in the treatment of schizophrenia.},
journal = {Journal of psychiatric research},
volume = {186},
number = {},
pages = {145-153},
doi = {10.1016/j.jpsychires.2025.04.019},
pmid = {40245529},
issn = {1879-1379},
abstract = {Ampakines, positive allosteric modulators of AMPA-glutamate receptors (AMPAR), have therapeutic implications in neuropsychiatric and neurological disorders in which AMPAR signaling is compromised such as Alzheimer's, ADHD, and schizophrenia. Low impact ampakines are a distinct subset of ampakines that only partially offset receptor desensitization and do not meaningfully alter binding affinity of AMPAR agonists, which may explain their lack of seizurogenic effects seen with other AMPAR positive modulators. Herein, we describe the preclinical pharmacology and antipsychotic activity of the low impact ampakine 1-(benzofurazan-5-ylcarbonyl)piperidine (CX691). CX691 moderately offsets desensitization in hippocampal patches, supporting its designation as a low impact ampakine and penetrates the blood-brain barrier. CX691 is more potent than well characterized high impact ampakines CX614 and CX546 and low impact ampakine CX516 in abrogating amphetamine-stimulated locomotor activity in Sprague Dawley rats. Low-dose CX691 synergistically reduces methamphetamine-induced locomotor activity when paired with approved antipsychotics clozapine and olanzapine. In rats treated chronically with amphetamine, CX691 retains antipsychotic activity whereas high doses of CX516 lack therapeutic effects. In contrast to haloperidol, CX691 is devoid of cataleptic activity at supratherapeutic doses in rats. CX691 enhances performance in the eight-arm radial maze, a spatial task that assesses hippocampal function, an activity of potential value for ameliorating cognitive deficits in schizophrenia. Taken together, these findings illustrate that low impact ampakines with improved potency might be useful therapeutic interventions in schizophrenic patients when given alone or as adjuncts to ongoing traditional antipsychotic drug therapies.},
}
RevDate: 2025-04-17
CmpDate: 2025-04-17
Generating Artificial Patients With Reliable Clinical Characteristics Using a Geometry-Based Variational Autoencoder: Proof-of-Concept Feasibility Study.
Journal of medical Internet research, 27:e63130 pii:v27i1e63130.
BACKGROUND: Artificial patient technology could transform health care by accelerating diagnosis, treatment, and mapping clinical pathways. Deep learning methods for generating artificial data in health care include data augmentation by variational autoencoders (VAE) technology.
OBJECTIVE: We aimed to test the feasibility of generating artificial patients with reliable clinical characteristics by using a geometry-based VAE applied, for the first time, on high-dimension, low-sample-size tabular data.
METHODS: Clinical tabular data were extracted from 521 real patients of the "MAX" digital conversational agent (BOTdesign) created for preparing patients for anesthesia. A 3-stage methodological approach was implemented to generate up to 10,000 artificial patients: training the model and generating artificial data, assessing the consistency and confidentiality of artificial data, and validating the plausibility of the newly created artificial patients.
RESULTS: We demonstrated the feasibility of applying the VAE technique to tabular data to generate large artificial patient cohorts with high consistency (fidelity scores>94%). Moreover, artificial patients could not be matched with real patients (filter similarity scores>99%, κ coefficients of agreement<0.2), thus guaranteeing the essential ethical concern of confidentiality.
CONCLUSIONS: This proof-of-concept study has demonstrated our ability to augment real tabular data to generate artificial patients. These promising results make it possible to envisage in silico trials carried out on large cohorts of artificial patients, thereby overcoming the pitfalls usually encountered in in vivo trials. Further studies integrating longitudinal dynamics are needed to map patient trajectories.
Additional Links: PMID-40245392
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40245392,
year = {2025},
author = {Ferré, F and Allassonnière, S and Chadebec, C and Minville, V},
title = {Generating Artificial Patients With Reliable Clinical Characteristics Using a Geometry-Based Variational Autoencoder: Proof-of-Concept Feasibility Study.},
journal = {Journal of medical Internet research},
volume = {27},
number = {},
pages = {e63130},
doi = {10.2196/63130},
pmid = {40245392},
issn = {1438-8871},
mesh = {Humans ; Feasibility Studies ; Proof of Concept Study ; *Deep Learning ; Autoencoder ; },
abstract = {BACKGROUND: Artificial patient technology could transform health care by accelerating diagnosis, treatment, and mapping clinical pathways. Deep learning methods for generating artificial data in health care include data augmentation by variational autoencoders (VAE) technology.
OBJECTIVE: We aimed to test the feasibility of generating artificial patients with reliable clinical characteristics by using a geometry-based VAE applied, for the first time, on high-dimension, low-sample-size tabular data.
METHODS: Clinical tabular data were extracted from 521 real patients of the "MAX" digital conversational agent (BOTdesign) created for preparing patients for anesthesia. A 3-stage methodological approach was implemented to generate up to 10,000 artificial patients: training the model and generating artificial data, assessing the consistency and confidentiality of artificial data, and validating the plausibility of the newly created artificial patients.
RESULTS: We demonstrated the feasibility of applying the VAE technique to tabular data to generate large artificial patient cohorts with high consistency (fidelity scores>94%). Moreover, artificial patients could not be matched with real patients (filter similarity scores>99%, κ coefficients of agreement<0.2), thus guaranteeing the essential ethical concern of confidentiality.
CONCLUSIONS: This proof-of-concept study has demonstrated our ability to augment real tabular data to generate artificial patients. These promising results make it possible to envisage in silico trials carried out on large cohorts of artificial patients, thereby overcoming the pitfalls usually encountered in in vivo trials. Further studies integrating longitudinal dynamics are needed to map patient trajectories.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Feasibility Studies
Proof of Concept Study
*Deep Learning
Autoencoder
RevDate: 2025-04-17
CmpDate: 2025-04-17
Biological Effects of Dietary Restriction on Alzheimer's Disease: Experimental and Clinical Investigations.
CNS neuroscience & therapeutics, 31(4):e70392.
BACKGROUNDS: Dementia can impose a heavy economic burden on both society and families. Alzheimer's disease (AD), the most prevalent form of dementia, is a complex neurodegenerative disease characterized by the abnormal deposition of extracellular amyloid β-protein (Aβ) and the aggregation of intracellular Tau protein to form neurofibrillary tangles (NFTs). Given the limited efficacy of pharmacological treatment, scientists have already paid more attention to non-pharmacological strategies, including dietary restriction (DR). DR refers to a nutritional paradigm aimed at promoting overall health by modifying the balance between energy consumption and expenditure. Studies have demonstrated that DR effectively extends the healthy lifespan, delays the aging process, and achieves promising results in the prevention and treatment of AD in preclinical studies.
METHODS: In this review we collected related studies and viewpoints by searching on PubMed database using the keywords. Most of the citations were published between 2015 and 2025. A few older literatures were also included due to their relevance and significance in this field.
RESULTS: We first provide a concise overview of the current therapeutic and preventive strategies for AD. Then, we introduce several specific DR protocols and their favorable effects on AD. Furthermore, the potential mechanisms underlying the benefits of DR on AD are discussed. Finally, we briefly highlight the role of DR in maintaining brain health.
CONCLUSION: This review may offer valuable insights into the development of innovative non-pharmacological strategies for AD treatment.
Additional Links: PMID-40245176
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40245176,
year = {2025},
author = {Liu, Z and Zhang, J and Jiang, F and Liu, C and Shao, Y and Le, W},
title = {Biological Effects of Dietary Restriction on Alzheimer's Disease: Experimental and Clinical Investigations.},
journal = {CNS neuroscience & therapeutics},
volume = {31},
number = {4},
pages = {e70392},
doi = {10.1111/cns.70392},
pmid = {40245176},
issn = {1755-5949},
support = {YDZX20213100001002//Shanghai Municipal Central Government Funds for Guiding Local Scientific and Technological Development/ ; XLYC2403127//Young Top Talents in the Xingliao Talents Program of Liaoning Province/ ; 32220103006//National Natural Science Foundation of China/ ; 82271524//National Natural Science Foundation of China/ ; 2024RY003//Dalian Science and Technology Talent Innovation Support Program/ ; },
mesh = {*Alzheimer Disease/diet therapy/metabolism ; Humans ; Animals ; *Caloric Restriction/methods ; Brain/metabolism ; },
abstract = {BACKGROUNDS: Dementia can impose a heavy economic burden on both society and families. Alzheimer's disease (AD), the most prevalent form of dementia, is a complex neurodegenerative disease characterized by the abnormal deposition of extracellular amyloid β-protein (Aβ) and the aggregation of intracellular Tau protein to form neurofibrillary tangles (NFTs). Given the limited efficacy of pharmacological treatment, scientists have already paid more attention to non-pharmacological strategies, including dietary restriction (DR). DR refers to a nutritional paradigm aimed at promoting overall health by modifying the balance between energy consumption and expenditure. Studies have demonstrated that DR effectively extends the healthy lifespan, delays the aging process, and achieves promising results in the prevention and treatment of AD in preclinical studies.
METHODS: In this review we collected related studies and viewpoints by searching on PubMed database using the keywords. Most of the citations were published between 2015 and 2025. A few older literatures were also included due to their relevance and significance in this field.
RESULTS: We first provide a concise overview of the current therapeutic and preventive strategies for AD. Then, we introduce several specific DR protocols and their favorable effects on AD. Furthermore, the potential mechanisms underlying the benefits of DR on AD are discussed. Finally, we briefly highlight the role of DR in maintaining brain health.
CONCLUSION: This review may offer valuable insights into the development of innovative non-pharmacological strategies for AD treatment.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/diet therapy/metabolism
Humans
Animals
*Caloric Restriction/methods
Brain/metabolism
RevDate: 2025-04-17
CmpDate: 2025-04-17
Evaluation of AccuBrain-based MRI quantitative analysis in diagnosing Alzheimer's disease and assessing behavioral and psychological symptoms of dementia.
Aging clinical and experimental research, 37(1):126.
AIM: Alzheimer's disease (AD) is a major cause of dementia, marked by cognitive decline and behavioral and psychological symptoms of dementia (BPSD). Early differentiation between AD, mild cognitive impairment (MCI), and healthy controls (HC) is essential for improving diagnostic accuracy and guiding effective treatment strategies.
METHODS: This retrospective study included 120 participants divided into AD (n = 40), MCI (n = 40), and HC (n = 40) groups. Brain MRI data were analyzed using the AccuBrain system to quantify AD Resemblance Atrophy Index (AD-RAI), Quantitative Medial Temporal Atrophy (QMTA), hippocampal volume, and white matter hyperintensities. Correlation analyses were conducted between imaging biomarkers and cognitive function scores (Mini-Mental State Examination, MMSE; Neuropsychiatric Inventory, NPI). Receiver operating characteristic (ROC) curve analysis was used to evaluated the diagnostic performance of the biomarkers.
RESULTS: AD patients had significantly higher AD-RAI (0.91 ± 0.25) and more pronounced hippocampal atrophy (0.36 ± 0.09) compared to MCI and HC (P < 0.001). Correlation analyses showed that AD-RAI and QMTA were negatively correlated with MMSE scores (r = -0.718, P < 0.001; r = -0.463, P < 0.001), while hippocampal volume was positively correlated with MMSE (r = 0.408, P < 0.001). ROC analysis revealed that AD-RAI had an AUC of 0.777 for distinguishing AD from MCI, while QMTA had an AUC of 0.938 for distinguishing AD from HC. BPSD patients exhibited higher AD-RAI (1.09 ± 0.18) and greater hippocampal atrophy, with ROC AUC > 0.9 for distinguishing BPSD from non-BPSD patients.
CONCLUSION: The AccuBrain MRI system demonstrated high sensitivity and diagnostic value in distinguishing AD from MCI and HC, as well as in identifying patients with BPSD. Correlation and ROC analyses support the use of these imaging biomarkers for early diagnosis and personalized treatment strategies in AD.
Additional Links: PMID-40244500
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40244500,
year = {2025},
author = {Lu, H and Xu, C and Liang, J},
title = {Evaluation of AccuBrain-based MRI quantitative analysis in diagnosing Alzheimer's disease and assessing behavioral and psychological symptoms of dementia.},
journal = {Aging clinical and experimental research},
volume = {37},
number = {1},
pages = {126},
pmid = {40244500},
issn = {1720-8319},
support = {2220001004730//the project of Foshan Science and Technology Bureau/ ; },
mesh = {Humans ; *Alzheimer Disease/diagnostic imaging/psychology/diagnosis ; Male ; Female ; *Magnetic Resonance Imaging/methods ; Aged ; Retrospective Studies ; *Cognitive Dysfunction/diagnostic imaging/diagnosis ; Aged, 80 and over ; ROC Curve ; Atrophy ; Hippocampus/diagnostic imaging/pathology ; Neuropsychological Tests ; Biomarkers ; Middle Aged ; *Dementia/psychology/diagnostic imaging ; },
abstract = {AIM: Alzheimer's disease (AD) is a major cause of dementia, marked by cognitive decline and behavioral and psychological symptoms of dementia (BPSD). Early differentiation between AD, mild cognitive impairment (MCI), and healthy controls (HC) is essential for improving diagnostic accuracy and guiding effective treatment strategies.
METHODS: This retrospective study included 120 participants divided into AD (n = 40), MCI (n = 40), and HC (n = 40) groups. Brain MRI data were analyzed using the AccuBrain system to quantify AD Resemblance Atrophy Index (AD-RAI), Quantitative Medial Temporal Atrophy (QMTA), hippocampal volume, and white matter hyperintensities. Correlation analyses were conducted between imaging biomarkers and cognitive function scores (Mini-Mental State Examination, MMSE; Neuropsychiatric Inventory, NPI). Receiver operating characteristic (ROC) curve analysis was used to evaluated the diagnostic performance of the biomarkers.
RESULTS: AD patients had significantly higher AD-RAI (0.91 ± 0.25) and more pronounced hippocampal atrophy (0.36 ± 0.09) compared to MCI and HC (P < 0.001). Correlation analyses showed that AD-RAI and QMTA were negatively correlated with MMSE scores (r = -0.718, P < 0.001; r = -0.463, P < 0.001), while hippocampal volume was positively correlated with MMSE (r = 0.408, P < 0.001). ROC analysis revealed that AD-RAI had an AUC of 0.777 for distinguishing AD from MCI, while QMTA had an AUC of 0.938 for distinguishing AD from HC. BPSD patients exhibited higher AD-RAI (1.09 ± 0.18) and greater hippocampal atrophy, with ROC AUC > 0.9 for distinguishing BPSD from non-BPSD patients.
CONCLUSION: The AccuBrain MRI system demonstrated high sensitivity and diagnostic value in distinguishing AD from MCI and HC, as well as in identifying patients with BPSD. Correlation and ROC analyses support the use of these imaging biomarkers for early diagnosis and personalized treatment strategies in AD.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alzheimer Disease/diagnostic imaging/psychology/diagnosis
Male
Female
*Magnetic Resonance Imaging/methods
Aged
Retrospective Studies
*Cognitive Dysfunction/diagnostic imaging/diagnosis
Aged, 80 and over
ROC Curve
Atrophy
Hippocampus/diagnostic imaging/pathology
Neuropsychological Tests
Biomarkers
Middle Aged
*Dementia/psychology/diagnostic imaging
RevDate: 2025-04-17
CmpDate: 2025-04-17
Oxymatrine impedes Alzheimer's progression via the attenuation of hypercholesterolemia and fibrosis.
Metabolic brain disease, 40(5):187.
This study highlights the potential therapeutic benefits of oxymatrine (OMT), a quinolizidine alkaloid found in Sophora flavescens, for Alzheimer's disease (AD). This study connects the dots between metabolic and neuronal origins by exploring the effects of oxymatrine in slowing down hypercholesterolemic and fibrotic changes that contribute to cognitive deficits. In our study, laboratory rats were fed a high-cholesterol diet for eight weeks. Cognitive abilities were assessed weekly using Hebb's Williams Maze and Radial arm mazes. Additionally, intraperitoneal doses of OMT were administered (20 mg/kg, 40 mg/kg, and 80 mg/kg) for 21 days. Furthermore, using ELISA, plasma and brain oxysterols, transforming growth factor β, amyloid β, matrix metalloproteinase- 9, claudin- 5, and ATP Binding Cassette Transporter A1 levels were measured biweekly. High-density lipoprotein, low-density lipoprotein, aspartate aminotransferase, and alanine transaminase levels were estimated using diagnostic kits. The findings demonstrate that The administration of oxymatrine to experimental animals resulted in a dose-dependent synergistic decline in several biomarkers, including oxysterols, transforming growth factor β, amyloid β, matrix metalloproteinase- 9, low-density lipoprotein, aspartate aminotransferase, and alanine transaminase levels. At the same time, a concomitant increase in the levels of Claudin- 5, ATP Binding Cassette transporter A1, high-density lipoprotein, and antioxidants in the same animals was observed, especially at a dose of 80 mg/kg. This study aims to establish a link between metabolic and neural origins by investigating the effects of oxymatrine in reducing the progression of hypercholesterolemia and fibrosis, which contribute to cognitive impairment in AD. The research explores how oxymatrine regulates mediators involved in oxysterol production and fibrotic alterations in AD. Preliminary results suggest that oxymatrine has the potential to significantly delay the development and progression of AD, offering a promising treatment alternative for those affected by the disease. The findings of the present study strongly suggest that OMT effectively retards the progression of AD, which is commonly associated with the intake of high-cholesterol diets. Subsequent investigations ought to examine the molecular mechanisms behind oxymatrine's interaction with oxysterols and lipid metabolism, including sophisticated imaging methodologies and metabolomics. Longitudinal studies are essential to evaluate the long-term efficacy and safety of oxymatrine in both animal models and people. Exploring its possible synergistic effects with current medications may yield more effective therapeutic techniques. Identifying biomarkers for personalised medication may also be beneficial. Clinical trials and research on oxymatrine's potential as a prophylactic medication may yield significant insights.
Additional Links: PMID-40244482
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40244482,
year = {2025},
author = {Das, A and Manna, R and Chowdhury, D and Sharma, D and Bodakhe, SH},
title = {Oxymatrine impedes Alzheimer's progression via the attenuation of hypercholesterolemia and fibrosis.},
journal = {Metabolic brain disease},
volume = {40},
number = {5},
pages = {187},
pmid = {40244482},
issn = {1573-7365},
mesh = {Animals ; *Alkaloids/pharmacology/therapeutic use ; *Alzheimer Disease/drug therapy/metabolism/pathology ; *Quinolizines/pharmacology/therapeutic use ; Rats ; Male ; *Hypercholesterolemia/drug therapy/metabolism ; Disease Progression ; Fibrosis/drug therapy/metabolism ; Brain/drug effects/metabolism/pathology ; Maze Learning/drug effects ; Rats, Wistar ; Matrines ; },
abstract = {This study highlights the potential therapeutic benefits of oxymatrine (OMT), a quinolizidine alkaloid found in Sophora flavescens, for Alzheimer's disease (AD). This study connects the dots between metabolic and neuronal origins by exploring the effects of oxymatrine in slowing down hypercholesterolemic and fibrotic changes that contribute to cognitive deficits. In our study, laboratory rats were fed a high-cholesterol diet for eight weeks. Cognitive abilities were assessed weekly using Hebb's Williams Maze and Radial arm mazes. Additionally, intraperitoneal doses of OMT were administered (20 mg/kg, 40 mg/kg, and 80 mg/kg) for 21 days. Furthermore, using ELISA, plasma and brain oxysterols, transforming growth factor β, amyloid β, matrix metalloproteinase- 9, claudin- 5, and ATP Binding Cassette Transporter A1 levels were measured biweekly. High-density lipoprotein, low-density lipoprotein, aspartate aminotransferase, and alanine transaminase levels were estimated using diagnostic kits. The findings demonstrate that The administration of oxymatrine to experimental animals resulted in a dose-dependent synergistic decline in several biomarkers, including oxysterols, transforming growth factor β, amyloid β, matrix metalloproteinase- 9, low-density lipoprotein, aspartate aminotransferase, and alanine transaminase levels. At the same time, a concomitant increase in the levels of Claudin- 5, ATP Binding Cassette transporter A1, high-density lipoprotein, and antioxidants in the same animals was observed, especially at a dose of 80 mg/kg. This study aims to establish a link between metabolic and neural origins by investigating the effects of oxymatrine in reducing the progression of hypercholesterolemia and fibrosis, which contribute to cognitive impairment in AD. The research explores how oxymatrine regulates mediators involved in oxysterol production and fibrotic alterations in AD. Preliminary results suggest that oxymatrine has the potential to significantly delay the development and progression of AD, offering a promising treatment alternative for those affected by the disease. The findings of the present study strongly suggest that OMT effectively retards the progression of AD, which is commonly associated with the intake of high-cholesterol diets. Subsequent investigations ought to examine the molecular mechanisms behind oxymatrine's interaction with oxysterols and lipid metabolism, including sophisticated imaging methodologies and metabolomics. Longitudinal studies are essential to evaluate the long-term efficacy and safety of oxymatrine in both animal models and people. Exploring its possible synergistic effects with current medications may yield more effective therapeutic techniques. Identifying biomarkers for personalised medication may also be beneficial. Clinical trials and research on oxymatrine's potential as a prophylactic medication may yield significant insights.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Alkaloids/pharmacology/therapeutic use
*Alzheimer Disease/drug therapy/metabolism/pathology
*Quinolizines/pharmacology/therapeutic use
Rats
Male
*Hypercholesterolemia/drug therapy/metabolism
Disease Progression
Fibrosis/drug therapy/metabolism
Brain/drug effects/metabolism/pathology
Maze Learning/drug effects
Rats, Wistar
Matrines
RevDate: 2025-04-17
CmpDate: 2025-04-17
A New Perspective on Agitation in Alzheimer's Disease: A Potential Paradigm Shift.
International journal of molecular sciences, 26(7): pii:ijms26073370.
Agitation is a common and difficult-to-manage neuropsychiatric syndrome in dementia. Recently, an association with the autonomous nervous system has been suggested. From the literature researched, however, only two studies investigating autonomic function concomitant to agitation situations appeared; one case series comprised two American veterans with vascular and Alzheimer's dementia, respectively, and in a case series of patients with CLN3 (juvenile neuronal ceroid lipofuscinosis), this was found to be the most common neurodegenerative disease leading to dementia in childhood. In both case series, the measurement of the autonomic system disclosed a parasympathetic withdrawal and sympathetic hyperactivity in the temporal context with agitated behavior. If the time-wise-related autonomic imbalance shown previously can be demonstrated in a larger cohort of patients with Alzheimer's disease, the use of transcutaneous vagal stimulation might be a potential paradigm shift in the treatment of agitation in Alzheimer's disease.
Additional Links: PMID-40244274
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40244274,
year = {2025},
author = {Ostergaard, JR},
title = {A New Perspective on Agitation in Alzheimer's Disease: A Potential Paradigm Shift.},
journal = {International journal of molecular sciences},
volume = {26},
number = {7},
pages = {},
doi = {10.3390/ijms26073370},
pmid = {40244274},
issn = {1422-0067},
mesh = {Humans ; *Alzheimer Disease/physiopathology/complications/therapy ; *Psychomotor Agitation/physiopathology/therapy/etiology ; Autonomic Nervous System/physiopathology ; Aged ; },
abstract = {Agitation is a common and difficult-to-manage neuropsychiatric syndrome in dementia. Recently, an association with the autonomous nervous system has been suggested. From the literature researched, however, only two studies investigating autonomic function concomitant to agitation situations appeared; one case series comprised two American veterans with vascular and Alzheimer's dementia, respectively, and in a case series of patients with CLN3 (juvenile neuronal ceroid lipofuscinosis), this was found to be the most common neurodegenerative disease leading to dementia in childhood. In both case series, the measurement of the autonomic system disclosed a parasympathetic withdrawal and sympathetic hyperactivity in the temporal context with agitated behavior. If the time-wise-related autonomic imbalance shown previously can be demonstrated in a larger cohort of patients with Alzheimer's disease, the use of transcutaneous vagal stimulation might be a potential paradigm shift in the treatment of agitation in Alzheimer's disease.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alzheimer Disease/physiopathology/complications/therapy
*Psychomotor Agitation/physiopathology/therapy/etiology
Autonomic Nervous System/physiopathology
Aged
RevDate: 2025-04-17
CmpDate: 2025-04-17
Repulsive Guidance Molecule-A as a Therapeutic Target Across Neurological Disorders: An Update.
International journal of molecular sciences, 26(7): pii:ijms26073221.
Repulsive guidance molecule-a (RGMa) has emerged as a significant therapeutic target in a variety of neurological disorders, including neurodegenerative diseases and acute conditions. This review comprehensively examines the multifaceted role of RGMa in central nervous system (CNS) pathologies such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, neuromyelitis optica spectrum disorder, spinal cord injury, stroke, vascular dementia, auditory neuropathy, and epilepsy. The mechanisms through which RGMa contributes to neuroinflammation, neuronal degeneration, and impaired axonal regeneration are herein discussed. Evidence from preclinical studies associate RGMa overexpression with negative outcomes, such as increased neuroinflammation and synaptic loss, while RGMa inhibition, particularly the use of agents like elezanumab, has shown promise in enhancing neuronal survival and functional recovery. RGMa's responses concerning immunomodulation and neurogenesis highlight its potential as a therapeutic avenue. We emphasize RGMa's critical role in CNS pathology and its potential to pave the way for innovative treatment strategies in neurological disorders. While preclinical findings are encouraging so far, further clinical trials are needed to validate the safety and efficacy of RGMa-targeted therapies.
Additional Links: PMID-40244061
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40244061,
year = {2025},
author = {Tseriotis, VS and Liampas, A and Lazaridou, IZ and Karachrysafi, S and Vavougios, GD and Hadjigeorgiou, GM and Papamitsou, T and Kouvelas, D and Arnaoutoglou, M and Pourzitaki, C and Mavridis, T},
title = {Repulsive Guidance Molecule-A as a Therapeutic Target Across Neurological Disorders: An Update.},
journal = {International journal of molecular sciences},
volume = {26},
number = {7},
pages = {},
doi = {10.3390/ijms26073221},
pmid = {40244061},
issn = {1422-0067},
mesh = {Humans ; Animals ; *Nervous System Diseases/metabolism/drug therapy ; *Nerve Tissue Proteins/metabolism/antagonists & inhibitors/genetics ; *GPI-Linked Proteins/metabolism/antagonists & inhibitors/genetics ; Molecular Targeted Therapy ; Neurodegenerative Diseases/metabolism/drug therapy ; },
abstract = {Repulsive guidance molecule-a (RGMa) has emerged as a significant therapeutic target in a variety of neurological disorders, including neurodegenerative diseases and acute conditions. This review comprehensively examines the multifaceted role of RGMa in central nervous system (CNS) pathologies such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, neuromyelitis optica spectrum disorder, spinal cord injury, stroke, vascular dementia, auditory neuropathy, and epilepsy. The mechanisms through which RGMa contributes to neuroinflammation, neuronal degeneration, and impaired axonal regeneration are herein discussed. Evidence from preclinical studies associate RGMa overexpression with negative outcomes, such as increased neuroinflammation and synaptic loss, while RGMa inhibition, particularly the use of agents like elezanumab, has shown promise in enhancing neuronal survival and functional recovery. RGMa's responses concerning immunomodulation and neurogenesis highlight its potential as a therapeutic avenue. We emphasize RGMa's critical role in CNS pathology and its potential to pave the way for innovative treatment strategies in neurological disorders. While preclinical findings are encouraging so far, further clinical trials are needed to validate the safety and efficacy of RGMa-targeted therapies.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Animals
*Nervous System Diseases/metabolism/drug therapy
*Nerve Tissue Proteins/metabolism/antagonists & inhibitors/genetics
*GPI-Linked Proteins/metabolism/antagonists & inhibitors/genetics
Molecular Targeted Therapy
Neurodegenerative Diseases/metabolism/drug therapy
RevDate: 2025-04-17
CmpDate: 2025-04-17
Cannabinerol Restores mRNA Splicing Defects Induced by β-Amyloid in an In Vitro Model of Alzheimer's Disease: A Transcriptomic Study.
International journal of molecular sciences, 26(7): pii:ijms26073113.
Alzheimer's disease (AD) is the most common form of dementia, characterized by β-amyloid (Aβ) plaques and neurofibrillary tangles, leading to neuronal loss and cognitive impairments. Recent studies have reported the dysregulation of RNA splicing in AD pathogenesis. Our previous transcriptomic study demonstrated the neuroprotective effect of the phytocannabinoid cannabinerol (CBNR) against the cell viability loss induced by Aβ in differentiated SH-SY5Y cells. This study also highlighted the deregulation of genes involved in mRNA splicing after Aβ exposure or CBNR pre-treatment. Here, we investigated whether CBNR could restore the splicing defects induced by Aβ in an AD in vitro model. Using the rMATS computational tool for detecting differential alternative splicing events (DASEs) from RNA-Seq data, we obtained 96 DASEs regulated in both conditions and, remarkably, they were all restored by CBNR pre-treatment. The pathway analysis indicated an over-representation of the "Alzheimer's disease-amyloid secretase pathway". Additionally, we observed that Aβ exposure increased the frequency of retained introns (RIs) among the shared DASEs, and that this frequency returned to normality by CBNR pre-treatment. Interestingly, most of these RIs contain a premature in-frame stop codon within the RNA sequence. Finally, analyzing the DASE regions for miRNA hybridization, we found 33 potential DASE/miRNA interactions that were relevant in AD pathogenesis. These findings revealed a novel trans-gene regulation by CBNR, potentially explaining part of its neuroprotective role. This is the first study demonstrating the involvement of a cannabinoid in the regulation of mRNA splicing in an AD model.
Additional Links: PMID-40243843
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40243843,
year = {2025},
author = {Lui, M and Salamone, S and Pollastro, F and Mazzon, E and Artimagnella, O},
title = {Cannabinerol Restores mRNA Splicing Defects Induced by β-Amyloid in an In Vitro Model of Alzheimer's Disease: A Transcriptomic Study.},
journal = {International journal of molecular sciences},
volume = {26},
number = {7},
pages = {},
doi = {10.3390/ijms26073113},
pmid = {40243843},
issn = {1422-0067},
support = {Current Research Funds 2024//Ministry of Health, Italy/ ; },
mesh = {Humans ; *Alzheimer Disease/genetics/metabolism/drug therapy/pathology ; *Amyloid beta-Peptides/metabolism ; *Transcriptome/drug effects ; *RNA, Messenger/genetics ; *Alternative Splicing/drug effects ; *RNA Splicing/drug effects ; Cell Line, Tumor ; Gene Expression Profiling ; },
abstract = {Alzheimer's disease (AD) is the most common form of dementia, characterized by β-amyloid (Aβ) plaques and neurofibrillary tangles, leading to neuronal loss and cognitive impairments. Recent studies have reported the dysregulation of RNA splicing in AD pathogenesis. Our previous transcriptomic study demonstrated the neuroprotective effect of the phytocannabinoid cannabinerol (CBNR) against the cell viability loss induced by Aβ in differentiated SH-SY5Y cells. This study also highlighted the deregulation of genes involved in mRNA splicing after Aβ exposure or CBNR pre-treatment. Here, we investigated whether CBNR could restore the splicing defects induced by Aβ in an AD in vitro model. Using the rMATS computational tool for detecting differential alternative splicing events (DASEs) from RNA-Seq data, we obtained 96 DASEs regulated in both conditions and, remarkably, they were all restored by CBNR pre-treatment. The pathway analysis indicated an over-representation of the "Alzheimer's disease-amyloid secretase pathway". Additionally, we observed that Aβ exposure increased the frequency of retained introns (RIs) among the shared DASEs, and that this frequency returned to normality by CBNR pre-treatment. Interestingly, most of these RIs contain a premature in-frame stop codon within the RNA sequence. Finally, analyzing the DASE regions for miRNA hybridization, we found 33 potential DASE/miRNA interactions that were relevant in AD pathogenesis. These findings revealed a novel trans-gene regulation by CBNR, potentially explaining part of its neuroprotective role. This is the first study demonstrating the involvement of a cannabinoid in the regulation of mRNA splicing in an AD model.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alzheimer Disease/genetics/metabolism/drug therapy/pathology
*Amyloid beta-Peptides/metabolism
*Transcriptome/drug effects
*RNA, Messenger/genetics
*Alternative Splicing/drug effects
*RNA Splicing/drug effects
Cell Line, Tumor
Gene Expression Profiling
RevDate: 2025-04-17
CmpDate: 2025-04-17
Neuroprotective Effects of Cilomilast and Chlorogenic Acid Against Scopolamine-Induced Memory Deficits via Modulation of the cAMP/PKA-CREB-BDNF Pathway.
International journal of molecular sciences, 26(7): pii:ijms26073108.
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline, neuroinflammation and neuronal damage. This study aimed to investigate the neuroprotective effects of cilomilast (CILO), a phosphodiesterase-4 (PDE4) inhibitor, alone and in combination with chlorogenic acid (CGA), a natural polyphenol, against scopolamine (SCOP)-induced cognitive impairment in mice. Forty male albino mice were divided into five groups: normal control, SCOP control, CGA + SCOP, CILO + SCOP and CILO + CGA + SCOP. Behavioral assessments, including the Y-maze and pole climbing tests, demonstrated that SCOP significantly impaired cognition, while treatment with CILO and CGA reversed these deficits, with the combination group showing the greatest improvement. Histopathological analyses revealed that CILO and CGA reduced neuronal damage and amyloid beta (Aβ) accumulation. Immunohistochemical and biochemical assessments confirmed a decrease in neuroinflammatory markers, including tumor necrosis factor-alpha (TNF-α) and nuclear factor kappa B (NF-κB). Molecular analyses showed that CILO restored cyclic adenosine monophosphate (cAMP) levels, leading to activation of protein kinase A (PKA), cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF), key regulators of neuronal plasticity and survival. CGA enhanced these effects by further inhibiting PDE4, amplifying the neuroprotective response. These findings suggest that PDE4 inhibitors, particularly in combination with CGA, may represent promising therapeutic strategies for AD-related cognitive impairment.
Additional Links: PMID-40243772
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40243772,
year = {2025},
author = {Mosalam, EM and Atya, SM and Mesbah, NM and Allam, S and Mehanna, ET},
title = {Neuroprotective Effects of Cilomilast and Chlorogenic Acid Against Scopolamine-Induced Memory Deficits via Modulation of the cAMP/PKA-CREB-BDNF Pathway.},
journal = {International journal of molecular sciences},
volume = {26},
number = {7},
pages = {},
doi = {10.3390/ijms26073108},
pmid = {40243772},
issn = {1422-0067},
mesh = {Animals ; Brain-Derived Neurotrophic Factor/metabolism ; *Neuroprotective Agents/pharmacology/therapeutic use ; Mice ; Scopolamine/adverse effects ; Male ; Cyclic AMP/metabolism ; *Memory Disorders/drug therapy/chemically induced/metabolism ; Cyclic AMP-Dependent Protein Kinases/metabolism ; *Chlorogenic Acid/pharmacology ; Cyclic AMP Response Element-Binding Protein/metabolism ; *Signal Transduction/drug effects ; Disease Models, Animal ; Phosphodiesterase 4 Inhibitors/pharmacology ; },
abstract = {Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline, neuroinflammation and neuronal damage. This study aimed to investigate the neuroprotective effects of cilomilast (CILO), a phosphodiesterase-4 (PDE4) inhibitor, alone and in combination with chlorogenic acid (CGA), a natural polyphenol, against scopolamine (SCOP)-induced cognitive impairment in mice. Forty male albino mice were divided into five groups: normal control, SCOP control, CGA + SCOP, CILO + SCOP and CILO + CGA + SCOP. Behavioral assessments, including the Y-maze and pole climbing tests, demonstrated that SCOP significantly impaired cognition, while treatment with CILO and CGA reversed these deficits, with the combination group showing the greatest improvement. Histopathological analyses revealed that CILO and CGA reduced neuronal damage and amyloid beta (Aβ) accumulation. Immunohistochemical and biochemical assessments confirmed a decrease in neuroinflammatory markers, including tumor necrosis factor-alpha (TNF-α) and nuclear factor kappa B (NF-κB). Molecular analyses showed that CILO restored cyclic adenosine monophosphate (cAMP) levels, leading to activation of protein kinase A (PKA), cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF), key regulators of neuronal plasticity and survival. CGA enhanced these effects by further inhibiting PDE4, amplifying the neuroprotective response. These findings suggest that PDE4 inhibitors, particularly in combination with CGA, may represent promising therapeutic strategies for AD-related cognitive impairment.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
Brain-Derived Neurotrophic Factor/metabolism
*Neuroprotective Agents/pharmacology/therapeutic use
Mice
Scopolamine/adverse effects
Male
Cyclic AMP/metabolism
*Memory Disorders/drug therapy/chemically induced/metabolism
Cyclic AMP-Dependent Protein Kinases/metabolism
*Chlorogenic Acid/pharmacology
Cyclic AMP Response Element-Binding Protein/metabolism
*Signal Transduction/drug effects
Disease Models, Animal
Phosphodiesterase 4 Inhibitors/pharmacology
RevDate: 2025-04-17
CmpDate: 2025-04-17
Oroxylum indicum (L.) Leaf Extract Attenuates β-Amyloid-Induced Neurotoxicity in SH-SY5Y Cells.
International journal of molecular sciences, 26(7): pii:ijms26072917.
Alzheimer's disease (AD) is characterized by the presence of amyloid-beta (Aβ) plaques, which trigger oxidative stress and neuronal cell death. The present study investigated the neuroprotective effects of Oroxylum indicum (L.) leaf (OIL) extract against Aβ-induced oxidative stress and cellular damage in SH-SY5Y cells. The cells were treated with OIL extract with and without Aβ25-35, and their viability was investigated. Moreover, the mechanism of action of OIL was assessed by determining caspase-3 levels, reactive oxygen species (ROS) and malondialdehyde (MDA) levels, enzymatic activity of catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px), phosphorylation of phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), extracellular signal-regulated kinase 1 and 2 (ERK1/2), and cAMP-responsive element-binding protein (CREB), and expression of B-cell lymphoma-2 (Bcl-2) proteins. The results indicated that OIL reduced Aβ-induced neurotoxicity in a concentration-dependent manner, improving cell viability, reducing ROS levels and MDA production, increasing antioxidant enzyme activity of CAT, SOD, and GSH-Px, and decreasing caspase-3 expression. In addition, OIL enhanced phosphorylation of Akt, ERK1/2, and CREB and upregulated Bcl-2 protein expression. High-performance liquid chromatography (HPLC) analysis identified oroxylin A, baicalein, and chrysin as the major phenolic constituents of the OIL extract. The findings suggest that the extract holds promise as a therapeutic intervention against Aβ-induced neurotoxicity, offering potential implications for the treatment of AD. Further studies are needed to investigate the activity of OIL in primary neurons or in vivo.
Additional Links: PMID-40243521
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40243521,
year = {2025},
author = {Palachai, N and Buranrat, B and Noisa, P and Mairuae, N},
title = {Oroxylum indicum (L.) Leaf Extract Attenuates β-Amyloid-Induced Neurotoxicity in SH-SY5Y Cells.},
journal = {International journal of molecular sciences},
volume = {26},
number = {7},
pages = {},
doi = {10.3390/ijms26072917},
pmid = {40243521},
issn = {1422-0067},
support = {//Thailand Science Research and Innovation/ ; },
mesh = {Humans ; *Plant Extracts/pharmacology/chemistry ; *Amyloid beta-Peptides/toxicity ; *Plant Leaves/chemistry ; *Neuroprotective Agents/pharmacology/chemistry ; Cell Line, Tumor ; Reactive Oxygen Species/metabolism ; Oxidative Stress/drug effects ; Cell Survival/drug effects ; *Bignoniaceae/chemistry ; Caspase 3/metabolism ; Antioxidants/pharmacology ; *Peptide Fragments/toxicity ; Neurons/drug effects/metabolism ; Malondialdehyde/metabolism ; },
abstract = {Alzheimer's disease (AD) is characterized by the presence of amyloid-beta (Aβ) plaques, which trigger oxidative stress and neuronal cell death. The present study investigated the neuroprotective effects of Oroxylum indicum (L.) leaf (OIL) extract against Aβ-induced oxidative stress and cellular damage in SH-SY5Y cells. The cells were treated with OIL extract with and without Aβ25-35, and their viability was investigated. Moreover, the mechanism of action of OIL was assessed by determining caspase-3 levels, reactive oxygen species (ROS) and malondialdehyde (MDA) levels, enzymatic activity of catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px), phosphorylation of phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), extracellular signal-regulated kinase 1 and 2 (ERK1/2), and cAMP-responsive element-binding protein (CREB), and expression of B-cell lymphoma-2 (Bcl-2) proteins. The results indicated that OIL reduced Aβ-induced neurotoxicity in a concentration-dependent manner, improving cell viability, reducing ROS levels and MDA production, increasing antioxidant enzyme activity of CAT, SOD, and GSH-Px, and decreasing caspase-3 expression. In addition, OIL enhanced phosphorylation of Akt, ERK1/2, and CREB and upregulated Bcl-2 protein expression. High-performance liquid chromatography (HPLC) analysis identified oroxylin A, baicalein, and chrysin as the major phenolic constituents of the OIL extract. The findings suggest that the extract holds promise as a therapeutic intervention against Aβ-induced neurotoxicity, offering potential implications for the treatment of AD. Further studies are needed to investigate the activity of OIL in primary neurons or in vivo.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Plant Extracts/pharmacology/chemistry
*Amyloid beta-Peptides/toxicity
*Plant Leaves/chemistry
*Neuroprotective Agents/pharmacology/chemistry
Cell Line, Tumor
Reactive Oxygen Species/metabolism
Oxidative Stress/drug effects
Cell Survival/drug effects
*Bignoniaceae/chemistry
Caspase 3/metabolism
Antioxidants/pharmacology
*Peptide Fragments/toxicity
Neurons/drug effects/metabolism
Malondialdehyde/metabolism
RevDate: 2025-04-17
CmpDate: 2025-04-17
Inhibitory Potential of Boscalid and Abamectin Towards Acetylcholinesterase and Butyrylcholinesterase: Computational and In Vitro Studies.
International journal of molecular sciences, 26(7): pii:ijms26072865.
The growing demand for agricultural products has led to the misuse of pesticides, resulting in the use of higher concentrations of these substances. This has led to an increase in toxicity imposed on other beneficial organisms and to the bioaccumulation of toxic pesticide concentrations in the bodies of both pests and non-target organisms, as well as in their end users, including humans. In this study, the neurotoxic potential of the commonly used pesticides abamectin (an insecticide) and boscalid (a fungicide) was evaluated. Both in vitro and in silico studies showed that human butyrylcholinesterase is not a target for abamectins B1A and B1B. Boscalid showed a modest Glide score (-28.8 kJ/mol) and a considerably higher IC50 (308.8 µM) against human butyrylcholinesterase than the approved inhibitor (2-((1-(benzenesulfonyl)-1H-indol-4-yl)oxy)ethyl)(benzyl)amine (IC50 = 0.473 µM). However, due to its non-mutagenicity and low toxicity, structural analogues of boscalid might be considered as candidates for the symptomatic treatment of Alzheimer's disease. Molecular dynamics simulations over 100 ns confirmed the stability of boscalid within the active site of butyrylcholinesterase, where it maintained key interactions with catalytic residues such as Trp82 and His438. These findings highlight its potential as a starting point for structure-based drug design strategies aimed at optimizing cholinesterase inhibitors with improved pharmacokinetic properties. According to absorption, distribution, metabolism, elimination, and toxicity studies, boscalid is orally active, which cannot be attributed to abamectins B1A and B1B.
Additional Links: PMID-40243451
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40243451,
year = {2025},
author = {Arsić, B and Petrović, S and Ilić, BS and Vrecl, M and Trobec, T and Sepčić, K and Frangež, R and Glišić, SM and Milićević, JS},
title = {Inhibitory Potential of Boscalid and Abamectin Towards Acetylcholinesterase and Butyrylcholinesterase: Computational and In Vitro Studies.},
journal = {International journal of molecular sciences},
volume = {26},
number = {7},
pages = {},
doi = {10.3390/ijms26072865},
pmid = {40243451},
issn = {1422-0067},
support = {337-00-21/2020-09/19//Ministry of Education, Science and Technological Development of the Republic of Serbia/ ; contract numbers 451-03-47/2023-01/200124 and 451-03-66/2024-03/200124 (B. Arsić, S. Petrović), and 451-03-47/2023-01/200017 and 451-03-66/2024-03/200017 (S. M. Glišić, J. S. Milićević)//Ministry of Science, Technological Development and Innovations of the Republic of Serbia/ ; research programs P4-0053 and P1-0207//Slovenian Research and innovation Agency/ ; },
mesh = {*Butyrylcholinesterase/chemistry/metabolism ; Humans ; *Ivermectin/analogs & derivatives/chemistry/pharmacology ; *Cholinesterase Inhibitors/pharmacology/chemistry ; *Acetylcholinesterase/chemistry/metabolism ; Molecular Dynamics Simulation ; Molecular Docking Simulation ; *Niacinamide/analogs & derivatives/chemistry/pharmacology ; },
abstract = {The growing demand for agricultural products has led to the misuse of pesticides, resulting in the use of higher concentrations of these substances. This has led to an increase in toxicity imposed on other beneficial organisms and to the bioaccumulation of toxic pesticide concentrations in the bodies of both pests and non-target organisms, as well as in their end users, including humans. In this study, the neurotoxic potential of the commonly used pesticides abamectin (an insecticide) and boscalid (a fungicide) was evaluated. Both in vitro and in silico studies showed that human butyrylcholinesterase is not a target for abamectins B1A and B1B. Boscalid showed a modest Glide score (-28.8 kJ/mol) and a considerably higher IC50 (308.8 µM) against human butyrylcholinesterase than the approved inhibitor (2-((1-(benzenesulfonyl)-1H-indol-4-yl)oxy)ethyl)(benzyl)amine (IC50 = 0.473 µM). However, due to its non-mutagenicity and low toxicity, structural analogues of boscalid might be considered as candidates for the symptomatic treatment of Alzheimer's disease. Molecular dynamics simulations over 100 ns confirmed the stability of boscalid within the active site of butyrylcholinesterase, where it maintained key interactions with catalytic residues such as Trp82 and His438. These findings highlight its potential as a starting point for structure-based drug design strategies aimed at optimizing cholinesterase inhibitors with improved pharmacokinetic properties. According to absorption, distribution, metabolism, elimination, and toxicity studies, boscalid is orally active, which cannot be attributed to abamectins B1A and B1B.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Butyrylcholinesterase/chemistry/metabolism
Humans
*Ivermectin/analogs & derivatives/chemistry/pharmacology
*Cholinesterase Inhibitors/pharmacology/chemistry
*Acetylcholinesterase/chemistry/metabolism
Molecular Dynamics Simulation
Molecular Docking Simulation
*Niacinamide/analogs & derivatives/chemistry/pharmacology
RevDate: 2025-04-17
CmpDate: 2025-04-17
Efficacy and Safety of Bilateral Deep Brain Stimulation (DBS) for Severe Alzheimer's Disease: A Comparative Analysis of Fornix Versus Basal Ganglia of Meynert.
CNS neuroscience & therapeutics, 31(4):e70285.
BACKGROUND: Deep brain stimulation (DBS) is a novel therapy for severe Alzheimer's disease (AD). However, there is an ongoing debate regarding the optimal target for DBS, particularly the fornix and the basal ganglia of Meynert (NBM).
OBJECTIVE: This study aimed to investigate the safety and efficacy of DBS for severe AD and to compare the fornix and the NBM as potential targets.
METHODS: We conducted a prospective, nonrandomized clinical study involving 20 patients with severe AD (MMSE score 0 to 10, CDR level 3) from January 2015 to August 2022, comprising 12 males and eight females, with a mean age of 59.05 ± 6.45 years. All patients underwent DBS treatment, among which 14 received bilateral fornix implantation, while six received bilateral implantation in the NBM. Electrical stimulation commenced 1 month postoperatively. We assessed the patients before surgery, followed by evaluations at 1 month, 3 months, 6 months, and 12 months poststimulation. Primary outcome measures focused on changes in cognitive function, assessed using the MMSE, MoCA, ADAS-Cog, and CDR scales. Secondary measures encompassed quality of life, caregiver burden, neuropsychiatric symptoms, and sleep disturbances, evaluated through the BI, FAQ, FIM, ZBI, NPI, HAMA, HAMD, and PSQI scales.
RESULTS: All patients tolerated DBS well, with no serious adverse effects reported. Early on, DBS significantly improved cognitive function and quality of life. Long-term benefits include the improvement of neuropsychiatric symptoms and sleep disorders and the alleviation of caregiver burden. Comparison between DBS targeting the NBM and fornix revealed no significant differences in overall scale scores. However, upon deeper analysis, NBM-DBS exhibited a more pronounced improvement in neuropsychiatric symptoms, particularly in NPI scores.
CONCLUSION: DBS is a potential therapeutic approach for severe AD, capable of improving patients' cognitive function, quality of life, and neuropsychiatric symptoms. Notably, NBM-DBS showed distinct advantages in ameliorating neuropsychiatric symptoms, providing valuable insights for clinically selecting the optimal DBS target.
TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT03115814.
Additional Links: PMID-40243219
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40243219,
year = {2025},
author = {Xu, J and Liu, B and Shang, G and Feng, Z and Yang, H and Chen, Y and Yu, X and Mao, Z},
title = {Efficacy and Safety of Bilateral Deep Brain Stimulation (DBS) for Severe Alzheimer's Disease: A Comparative Analysis of Fornix Versus Basal Ganglia of Meynert.},
journal = {CNS neuroscience & therapeutics},
volume = {31},
number = {4},
pages = {e70285},
doi = {10.1111/cns.70285},
pmid = {40243219},
issn = {1755-5949},
support = {2021ZD0200407//STI 2030-Major Projects/ ; },
mesh = {Humans ; *Deep Brain Stimulation/methods/adverse effects ; Male ; Female ; *Alzheimer Disease/therapy/psychology ; *Fornix, Brain/physiology ; Aged ; Middle Aged ; Prospective Studies ; Treatment Outcome ; *Basal Nucleus of Meynert/physiology ; },
abstract = {BACKGROUND: Deep brain stimulation (DBS) is a novel therapy for severe Alzheimer's disease (AD). However, there is an ongoing debate regarding the optimal target for DBS, particularly the fornix and the basal ganglia of Meynert (NBM).
OBJECTIVE: This study aimed to investigate the safety and efficacy of DBS for severe AD and to compare the fornix and the NBM as potential targets.
METHODS: We conducted a prospective, nonrandomized clinical study involving 20 patients with severe AD (MMSE score 0 to 10, CDR level 3) from January 2015 to August 2022, comprising 12 males and eight females, with a mean age of 59.05 ± 6.45 years. All patients underwent DBS treatment, among which 14 received bilateral fornix implantation, while six received bilateral implantation in the NBM. Electrical stimulation commenced 1 month postoperatively. We assessed the patients before surgery, followed by evaluations at 1 month, 3 months, 6 months, and 12 months poststimulation. Primary outcome measures focused on changes in cognitive function, assessed using the MMSE, MoCA, ADAS-Cog, and CDR scales. Secondary measures encompassed quality of life, caregiver burden, neuropsychiatric symptoms, and sleep disturbances, evaluated through the BI, FAQ, FIM, ZBI, NPI, HAMA, HAMD, and PSQI scales.
RESULTS: All patients tolerated DBS well, with no serious adverse effects reported. Early on, DBS significantly improved cognitive function and quality of life. Long-term benefits include the improvement of neuropsychiatric symptoms and sleep disorders and the alleviation of caregiver burden. Comparison between DBS targeting the NBM and fornix revealed no significant differences in overall scale scores. However, upon deeper analysis, NBM-DBS exhibited a more pronounced improvement in neuropsychiatric symptoms, particularly in NPI scores.
CONCLUSION: DBS is a potential therapeutic approach for severe AD, capable of improving patients' cognitive function, quality of life, and neuropsychiatric symptoms. Notably, NBM-DBS showed distinct advantages in ameliorating neuropsychiatric symptoms, providing valuable insights for clinically selecting the optimal DBS target.
TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT03115814.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Deep Brain Stimulation/methods/adverse effects
Male
Female
*Alzheimer Disease/therapy/psychology
*Fornix, Brain/physiology
Aged
Middle Aged
Prospective Studies
Treatment Outcome
*Basal Nucleus of Meynert/physiology
RevDate: 2025-04-17
Generating real-world evidence in early Alzheimer's disease: Considerations for applying the target trial emulation framework to study the safety of anti-amyloid therapies.
Alzheimer's & dementia (New York, N. Y.), 11(2):e70080.
UNLABELLED: Anti-amyloid beta monoclonal antibodies (anti-Aβ mAbs) have received approval from the US Food and Drug Administration for the treatment of patients with mild cognitive impairment or mild dementia due to Alzheimer's disease (collectively known as early AD) based on evidence from clinical trials. However, whether findings from these trials are generalizable to the real world is uncertain. We need reliable evidence on the real-world safety of these treatments to inform decision making for clinicians, patients, and caregivers. Using lecanemab as an exemplar, we outline the key considerations in designing and implementing an observational study on safety and utilization outcomes using established administrative healthcare claims data sources with the target trial emulation framework. The target trial emulation framework is a rigorous causal inference framework that minimizes common biases in observational studies. The approach proposed here can be applied to evaluation of additional mAbs as they become available.
HIGHLIGHTS: Little is known about real-world safety of anti-amyloid beta monoclonal antibodies for early Alzheimer's disease.Existing real-world data can support studies of their safety and utilization outcomes.Target trial emulation can guide the design of these studies while minimizing bias.We provide key design and analytical considerations for future studies.
Additional Links: PMID-40242567
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40242567,
year = {2025},
author = {Li, X and Singh, S and Rasouli, B and Lyons, J and Cocoros, NM and Platt, R and Abi-Elias, I and Gurwitz, JH},
title = {Generating real-world evidence in early Alzheimer's disease: Considerations for applying the target trial emulation framework to study the safety of anti-amyloid therapies.},
journal = {Alzheimer's & dementia (New York, N. Y.)},
volume = {11},
number = {2},
pages = {e70080},
pmid = {40242567},
issn = {2352-8737},
abstract = {UNLABELLED: Anti-amyloid beta monoclonal antibodies (anti-Aβ mAbs) have received approval from the US Food and Drug Administration for the treatment of patients with mild cognitive impairment or mild dementia due to Alzheimer's disease (collectively known as early AD) based on evidence from clinical trials. However, whether findings from these trials are generalizable to the real world is uncertain. We need reliable evidence on the real-world safety of these treatments to inform decision making for clinicians, patients, and caregivers. Using lecanemab as an exemplar, we outline the key considerations in designing and implementing an observational study on safety and utilization outcomes using established administrative healthcare claims data sources with the target trial emulation framework. The target trial emulation framework is a rigorous causal inference framework that minimizes common biases in observational studies. The approach proposed here can be applied to evaluation of additional mAbs as they become available.
HIGHLIGHTS: Little is known about real-world safety of anti-amyloid beta monoclonal antibodies for early Alzheimer's disease.Existing real-world data can support studies of their safety and utilization outcomes.Target trial emulation can guide the design of these studies while minimizing bias.We provide key design and analytical considerations for future studies.},
}
RevDate: 2025-04-17
A real-world disproportionality analysis of FDA adverse event reporting system (FAERS) events for lecanemab.
Frontiers in pharmacology, 16:1559447.
BACKGROUND: Lecanemab is a humanized murine IgG1 antibody. Recent Phase 3 clinical trials have demonstrated its ability to reduce brain amyloid-β (Aβ) load and slow cognitive decline in patients with early Alzheimer's disease (AD). However, since its approval, reports on adverse effects (AEs) associated with lecanemab have been limited. To better understand the AEs related to lecanemab and provide guidance for future clinical use, we analyzed lecanemab-associated AEs using data from the United States Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS).
METHODS: We extracted all AEs reports from the FAERS database for the period from the first quarter of 2023 to the third quarter of 2024. Using the Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and Multi-item Gamma Poisson Shrinker (MGPS) algorithms, we conducted a comprehensive analysis of lecanemab-related AEs, restricting the analysis to AEs with the role code of primary suspect (PS).
RESULTS: A total of 811 AEs reports related to lecanemab used in AD patients and 506 AEs in Non-AD patients were included. The preferred terms (PTs) identified as positive across all four algorithms included headache, Amyloid Related Imaging Abnormalities-oedema/effusion (ARIA-E), chills, Amyloid Related Imaging Abnormalities-haemosiderosis/microhaemorrhage (ARIA-H), fatigue, infusion-related reaction, nausea, pyrexia, pain, influenza like illness, and so on. Among these, ARIA-E, ARIA-H, brain oedema and status epilepticus were associated with Important Medical Events (IMEs) for AD patients, and brain oedema, cerebral haemorrhage, cerebral microhaemorrhage, subdural haematoma, ischaemic stroke, cerebral infarction were associated with IMEs for Non-AD patients. At the system organ class (SOC) level, the highest signal detection for lecanemab was observed in nervous system disorders among AD and Non-AD patients [ROR for AD: 2.42 (2.2-2.65); ROR for Non-AD: 6.97 (6.12-7.95)]. The median time to the occurrence of these AEs was 44 days after administration in AD patients and 30 days for Non-AD patients.
CONCLUSION: This study utilized the FAERS database to evaluate lecanemab-associated AEs in AD and non-AD patients, along with their temporal patterns post-marketing authorization, thereby establishing a foundation for subsequent clinical pharmacovigilance. A biweekly 10 mg/kg was identified as the optimal therapeutic dosage. ARIA emerged as frequent treatment-related AEs, with APOEɛ4 carriers demonstrating heightened susceptibility. This necessitates serial brain MRI surveillance for all patients during treatment, aimed not only at early ARIA detection but also vigilant monitoring of IMEs including cerebral haemorrhage, cerebral microhaemorrhages, subdural haematoma, cerebral edema, ischaemic stroke, and cerebral infarction. While AD patients predominantly exhibited non-specific clinical manifestations, non-AD cohorts showed elevated risks of stroke-related complications. Consequently, dynamic neurological deficit monitoring is indispensable for non-AD populations receiving lecanemab to mitigate adverse outcomes. Finally, comprehensive reassessment of anticoagulant or antiplatelet therapy indications is warranted in both AD and non-AD patients to reduce hemorrhagic risks.
Additional Links: PMID-40242445
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40242445,
year = {2025},
author = {Yan, L and Zhang, L and Xu, Z and Luo, Z},
title = {A real-world disproportionality analysis of FDA adverse event reporting system (FAERS) events for lecanemab.},
journal = {Frontiers in pharmacology},
volume = {16},
number = {},
pages = {1559447},
pmid = {40242445},
issn = {1663-9812},
abstract = {BACKGROUND: Lecanemab is a humanized murine IgG1 antibody. Recent Phase 3 clinical trials have demonstrated its ability to reduce brain amyloid-β (Aβ) load and slow cognitive decline in patients with early Alzheimer's disease (AD). However, since its approval, reports on adverse effects (AEs) associated with lecanemab have been limited. To better understand the AEs related to lecanemab and provide guidance for future clinical use, we analyzed lecanemab-associated AEs using data from the United States Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS).
METHODS: We extracted all AEs reports from the FAERS database for the period from the first quarter of 2023 to the third quarter of 2024. Using the Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and Multi-item Gamma Poisson Shrinker (MGPS) algorithms, we conducted a comprehensive analysis of lecanemab-related AEs, restricting the analysis to AEs with the role code of primary suspect (PS).
RESULTS: A total of 811 AEs reports related to lecanemab used in AD patients and 506 AEs in Non-AD patients were included. The preferred terms (PTs) identified as positive across all four algorithms included headache, Amyloid Related Imaging Abnormalities-oedema/effusion (ARIA-E), chills, Amyloid Related Imaging Abnormalities-haemosiderosis/microhaemorrhage (ARIA-H), fatigue, infusion-related reaction, nausea, pyrexia, pain, influenza like illness, and so on. Among these, ARIA-E, ARIA-H, brain oedema and status epilepticus were associated with Important Medical Events (IMEs) for AD patients, and brain oedema, cerebral haemorrhage, cerebral microhaemorrhage, subdural haematoma, ischaemic stroke, cerebral infarction were associated with IMEs for Non-AD patients. At the system organ class (SOC) level, the highest signal detection for lecanemab was observed in nervous system disorders among AD and Non-AD patients [ROR for AD: 2.42 (2.2-2.65); ROR for Non-AD: 6.97 (6.12-7.95)]. The median time to the occurrence of these AEs was 44 days after administration in AD patients and 30 days for Non-AD patients.
CONCLUSION: This study utilized the FAERS database to evaluate lecanemab-associated AEs in AD and non-AD patients, along with their temporal patterns post-marketing authorization, thereby establishing a foundation for subsequent clinical pharmacovigilance. A biweekly 10 mg/kg was identified as the optimal therapeutic dosage. ARIA emerged as frequent treatment-related AEs, with APOEɛ4 carriers demonstrating heightened susceptibility. This necessitates serial brain MRI surveillance for all patients during treatment, aimed not only at early ARIA detection but also vigilant monitoring of IMEs including cerebral haemorrhage, cerebral microhaemorrhages, subdural haematoma, cerebral edema, ischaemic stroke, and cerebral infarction. While AD patients predominantly exhibited non-specific clinical manifestations, non-AD cohorts showed elevated risks of stroke-related complications. Consequently, dynamic neurological deficit monitoring is indispensable for non-AD populations receiving lecanemab to mitigate adverse outcomes. Finally, comprehensive reassessment of anticoagulant or antiplatelet therapy indications is warranted in both AD and non-AD patients to reduce hemorrhagic risks.},
}
RevDate: 2025-04-17
GABAB Receptor: Structure, Biological Functions, and Therapy for Diseases.
MedComm, 6(5):e70163.
Gamma-aminobutyric acid (GABA) B receptors (GABABRs) that acts slowly and maintains the inhibitory tone are versatile regulators in the complex nervous behaviors and their involvement in various neuropsychiatric disorders, such as anxiety, epilepsy, pain, drug addiction, and Alzheimer's disease. Additional study advances have implied the crucial roles of GABABRs in regulating feeding-related behaviors, yet their therapeutic potential in addressing the neuropsychiatric disorders, binge eating, and feeding-related disorders remains underutilized. This general review summarized the physiological structure and functions of GABABR, explored the regulation in various psychiatric disorders, feeding behaviors, binge eating, and metabolism disorders, and fully discussed the potential of targeting GABABRs and its regulator-binding sites for the treatment of different psychiatric disorders, binge eating and even obesity. While agonists that directly bind to GABABR1 have some negative side effects, positive allosteric modulators (PAMs) that bind to GABABR2 demonstrate excellent therapeutic efficacy and tolerability and have better safety and therapeutic indexes. Moreover, phosphorylation sites of downstream GABABRs regulators may be novel therapeutic targets for psychiatric disorders, binge eating, and obesity. Further studies, clinical trials in particular, will be essential for confirming the therapeutic value of PAMs and other agents targeting the GABABR pathways in a clinical setting.
Additional Links: PMID-40242161
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40242161,
year = {2025},
author = {Xie, W and Li, Y and Wang, X and Blokhina, E and Krupitsky, E and Vetrova, M and Hu, J and Yuan, TF and Chen, J and Wang, H and Chen, X},
title = {GABAB Receptor: Structure, Biological Functions, and Therapy for Diseases.},
journal = {MedComm},
volume = {6},
number = {5},
pages = {e70163},
pmid = {40242161},
issn = {2688-2663},
abstract = {Gamma-aminobutyric acid (GABA) B receptors (GABABRs) that acts slowly and maintains the inhibitory tone are versatile regulators in the complex nervous behaviors and their involvement in various neuropsychiatric disorders, such as anxiety, epilepsy, pain, drug addiction, and Alzheimer's disease. Additional study advances have implied the crucial roles of GABABRs in regulating feeding-related behaviors, yet their therapeutic potential in addressing the neuropsychiatric disorders, binge eating, and feeding-related disorders remains underutilized. This general review summarized the physiological structure and functions of GABABR, explored the regulation in various psychiatric disorders, feeding behaviors, binge eating, and metabolism disorders, and fully discussed the potential of targeting GABABRs and its regulator-binding sites for the treatment of different psychiatric disorders, binge eating and even obesity. While agonists that directly bind to GABABR1 have some negative side effects, positive allosteric modulators (PAMs) that bind to GABABR2 demonstrate excellent therapeutic efficacy and tolerability and have better safety and therapeutic indexes. Moreover, phosphorylation sites of downstream GABABRs regulators may be novel therapeutic targets for psychiatric disorders, binge eating, and obesity. Further studies, clinical trials in particular, will be essential for confirming the therapeutic value of PAMs and other agents targeting the GABABR pathways in a clinical setting.},
}
RevDate: 2025-04-16
CmpDate: 2025-04-17
Impact of nucleos(t)ide analogue therapy on the incidence of Alzheimer's disease in patients with chronic hepatitis B virus infection.
Alzheimer's research & therapy, 17(1):84.
BACKGROUND: Long-term therapy with nucleos(t)ide analogs (NUCs) is inevitable for chronic hepatitis B (CHB) patients. However, how NUC therapy on the developing Alzheimer's disease (AD) in these patients remains controversial.
METHODS: This retrospective cohort study used the Korean National Health Insurance Service claims database from January 1, 2013, to December 31, 2013, treatment naïve CHB patients and those without previously diagnosed with AD. Participants were followed from the index date until either the diagnosis of AD or the study's conclusion on December 31, 2021. The primary outcome was the incidence of AD, compared between the group with initiated NUC therapy (n = 18,365) at cohort entry and the group without NUC therapy (n = 212,820).
RESULTS: During the study, 416 patients were diagnosed with AD. After propensity-score matching (18,365 pairs), the 5- to 7-year follow-up showed a significantly lower hazard ratio (HR) in the NUC-treated group compared to the untreated group (HR 0.31-0.40), with HRs remaining constant over time. Subgroup analysis showed more pronounced benefits of NUC therapy in patients under 65 years (HRs: 0.22 vs. 1.23; P < 0.05) and those without dyslipidemia (HRs: 0.14 vs. 1.09; P < 0.05). Protective effects were also observed across subgroups with hypertension, chronic kidney disease, heart disease, and a history of brain trauma, consistent with AD risk factor trends.
CONCLUSIONS: Our study analyses suggest that NUC therapy appears to have a protective effect against the development of AD in patients with CHB.
Additional Links: PMID-40241196
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40241196,
year = {2025},
author = {Lim, J and Gu, H and Sang, H and Jeong, SJ and Kim, HI},
title = {Impact of nucleos(t)ide analogue therapy on the incidence of Alzheimer's disease in patients with chronic hepatitis B virus infection.},
journal = {Alzheimer's research & therapy},
volume = {17},
number = {1},
pages = {84},
pmid = {40241196},
issn = {1758-9193},
support = {2023//The Korean Association for the Study of the Liver and The Korean Liver Foundation/ ; },
mesh = {Humans ; *Alzheimer Disease/epidemiology/prevention & control ; Male ; Female ; *Hepatitis B, Chronic/drug therapy/epidemiology ; Retrospective Studies ; Incidence ; Middle Aged ; Aged ; *Antiviral Agents/therapeutic use ; Republic of Korea/epidemiology ; *Nucleosides/therapeutic use ; Adult ; Cohort Studies ; },
abstract = {BACKGROUND: Long-term therapy with nucleos(t)ide analogs (NUCs) is inevitable for chronic hepatitis B (CHB) patients. However, how NUC therapy on the developing Alzheimer's disease (AD) in these patients remains controversial.
METHODS: This retrospective cohort study used the Korean National Health Insurance Service claims database from January 1, 2013, to December 31, 2013, treatment naïve CHB patients and those without previously diagnosed with AD. Participants were followed from the index date until either the diagnosis of AD or the study's conclusion on December 31, 2021. The primary outcome was the incidence of AD, compared between the group with initiated NUC therapy (n = 18,365) at cohort entry and the group without NUC therapy (n = 212,820).
RESULTS: During the study, 416 patients were diagnosed with AD. After propensity-score matching (18,365 pairs), the 5- to 7-year follow-up showed a significantly lower hazard ratio (HR) in the NUC-treated group compared to the untreated group (HR 0.31-0.40), with HRs remaining constant over time. Subgroup analysis showed more pronounced benefits of NUC therapy in patients under 65 years (HRs: 0.22 vs. 1.23; P < 0.05) and those without dyslipidemia (HRs: 0.14 vs. 1.09; P < 0.05). Protective effects were also observed across subgroups with hypertension, chronic kidney disease, heart disease, and a history of brain trauma, consistent with AD risk factor trends.
CONCLUSIONS: Our study analyses suggest that NUC therapy appears to have a protective effect against the development of AD in patients with CHB.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alzheimer Disease/epidemiology/prevention & control
Male
Female
*Hepatitis B, Chronic/drug therapy/epidemiology
Retrospective Studies
Incidence
Middle Aged
Aged
*Antiviral Agents/therapeutic use
Republic of Korea/epidemiology
*Nucleosides/therapeutic use
Adult
Cohort Studies
RevDate: 2025-04-17
The future of Alzheimer's treatment.
Nature, 640(8059):S4-S6.
Additional Links: PMID-40240849
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40240849,
year = {2025},
author = {Makin, S},
title = {The future of Alzheimer's treatment.},
journal = {Nature},
volume = {640},
number = {8059},
pages = {S4-S6},
pmid = {40240849},
issn = {1476-4687},
}
RevDate: 2025-04-16
The unusual genetic inheritance that could change Alzheimer's treatment.
Nature, 640(8059):S7.
Additional Links: PMID-40240844
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40240844,
year = {2025},
author = {Vargas-Parada, L},
title = {The unusual genetic inheritance that could change Alzheimer's treatment.},
journal = {Nature},
volume = {640},
number = {8059},
pages = {S7},
pmid = {40240844},
issn = {1476-4687},
}
RevDate: 2025-04-16
CmpDate: 2025-04-16
Development of donepezil hydrochloride-loaded PLGA-based nanoparticles for Alzheimer's disease treatment.
Scientific reports, 15(1):13184.
In recent years, nanoparticle (NP) systems have demonstrated significant promise in pharmaceutical applications. This study focused on the development of donepezil hydrochloride-loaded PLGA-NPs, prepared using the 'Double Emulsion Solvent Evaporation' method. The impact of varying concentrations of polyvinyl alcohol-(PVA) in the aqueous phase and sonication time on NP characteristics was comprehensively examined. Results showed that increasing PVA concentration and sonication time resulted in a reduction in NP size, with an optimal formulation (I-DNP) achieving a particle size of 136.37 nm ± 0.93 and a PDI of 0.122 ± 0.011, indicating uniformity. The zeta potential was measured at - 24.17mV ± 1.21, confirming the electrostatic stability of the formulation, essential for long-term stability. Trehalose was incorporated to enhance stability, and gastrointestinal stability testing revealed that I-DNP degraded faster in acidic environments. The encapsulation efficiency reached 69.22 ± 4.84%, suggesting effective drug loading, and release studies exhibited a sustained release profile, with a Fickian and non-Fickian release mechanism. DSC, FT-IR, and [1]H-NMR analyses confirmed the encapsulation and structural integrity of the formulation. In biological activity studies, I-DNP exhibited potent anti-AChE and anti-BuChE activities, with Chorioallantoic Membrane (CAM) assays showing significant inhibition of angiogenesis. These findings highlight the potential of I-DNP as a promising therapeutic strategy for Alzheimer's disease, demonstrating its ability to enhance drug stability, controlled release, and potential blood-brain barrier (BBB) penetration. Future studies will focus on long-term stability testing and in vivo Alzheimer's models to further validate its clinical applicability. This research contributes to the advancement of nanoparticle-based drug delivery systems for neurodegenerative diseases, paving the way for innovative therapeutic approaches.
Additional Links: PMID-40240764
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40240764,
year = {2025},
author = {Kömür, M and Kıyan, HT and Öztürk, AA},
title = {Development of donepezil hydrochloride-loaded PLGA-based nanoparticles for Alzheimer's disease treatment.},
journal = {Scientific reports},
volume = {15},
number = {1},
pages = {13184},
pmid = {40240764},
issn = {2045-2322},
support = {Project number: 2304S027, Project ID: 1956//Anadolu University Scientific Research Project Commission/ ; },
mesh = {*Donepezil/chemistry/pharmacology/administration & dosage ; *Alzheimer Disease/drug therapy ; *Nanoparticles/chemistry ; *Polylactic Acid-Polyglycolic Acid Copolymer/chemistry ; Drug Liberation ; Particle Size ; Animals ; Drug Carriers/chemistry ; Humans ; Chick Embryo ; },
abstract = {In recent years, nanoparticle (NP) systems have demonstrated significant promise in pharmaceutical applications. This study focused on the development of donepezil hydrochloride-loaded PLGA-NPs, prepared using the 'Double Emulsion Solvent Evaporation' method. The impact of varying concentrations of polyvinyl alcohol-(PVA) in the aqueous phase and sonication time on NP characteristics was comprehensively examined. Results showed that increasing PVA concentration and sonication time resulted in a reduction in NP size, with an optimal formulation (I-DNP) achieving a particle size of 136.37 nm ± 0.93 and a PDI of 0.122 ± 0.011, indicating uniformity. The zeta potential was measured at - 24.17mV ± 1.21, confirming the electrostatic stability of the formulation, essential for long-term stability. Trehalose was incorporated to enhance stability, and gastrointestinal stability testing revealed that I-DNP degraded faster in acidic environments. The encapsulation efficiency reached 69.22 ± 4.84%, suggesting effective drug loading, and release studies exhibited a sustained release profile, with a Fickian and non-Fickian release mechanism. DSC, FT-IR, and [1]H-NMR analyses confirmed the encapsulation and structural integrity of the formulation. In biological activity studies, I-DNP exhibited potent anti-AChE and anti-BuChE activities, with Chorioallantoic Membrane (CAM) assays showing significant inhibition of angiogenesis. These findings highlight the potential of I-DNP as a promising therapeutic strategy for Alzheimer's disease, demonstrating its ability to enhance drug stability, controlled release, and potential blood-brain barrier (BBB) penetration. Future studies will focus on long-term stability testing and in vivo Alzheimer's models to further validate its clinical applicability. This research contributes to the advancement of nanoparticle-based drug delivery systems for neurodegenerative diseases, paving the way for innovative therapeutic approaches.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Donepezil/chemistry/pharmacology/administration & dosage
*Alzheimer Disease/drug therapy
*Nanoparticles/chemistry
*Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
Drug Liberation
Particle Size
Animals
Drug Carriers/chemistry
Humans
Chick Embryo
RevDate: 2025-04-16
CmpDate: 2025-04-16
Unravelling the Proteinopathic Engagement of α-Synuclein, Tau, and Amyloid Beta in Parkinson's Disease: Mitochondrial Collapse as a Pivotal Driver of Neurodegeneration.
Neurochemical research, 50(3):145.
Parkinson's disease is a complex neurological ailment manifested by dopaminergic neurodegeneration in the substantia nigra of the brain. This study investigates the molecular tripartite interaction between Lewy bodies, amyloid beta, and tau protein in the pathogenesis of Parkinson's disease. Lewy bodies which have been found as the important pathological hallmark in the degenerative neurons of Parkinson's patients, are mainly composed of α-synuclein. The accumulation of α-synuclein has been directly and indirectly linked to the severity and degree of progression of the disease. In addition, approximately 50% of Parkinson's disease cases are also described by hyperphosphorylation of tau protein indicating its significant involvement in the disease. The study further explains how α-synuclein, tau and amyloid beta can spread via cross-seeding mechanisms and accelerate each other's aggregation leading to neuronal death. Both GSK-3β and CDK5 are involved in phosphorylation which among other effects contributes to the misfolding of both α-synuclein and tau proteins that lead to neurodegeneration in Alzheimer's disease. Several mediators, that contribute to mitochondrial damage through elevated oxidative stress pathology are clearly described. Because of the increase in the incidence of Parkinson's disease, as predicted to be 17 million when the study was being conducted, studying these pathological mechanisms is very important in trying to establish treatments. This work contributes a path to finding a multi-target treatment regimen to alleviate the burden of this devastating disease.
Additional Links: PMID-40240583
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40240583,
year = {2025},
author = {Sahoo, TA and Chand, J and Kandy, AT and Antony, S and Subramanian, G},
title = {Unravelling the Proteinopathic Engagement of α-Synuclein, Tau, and Amyloid Beta in Parkinson's Disease: Mitochondrial Collapse as a Pivotal Driver of Neurodegeneration.},
journal = {Neurochemical research},
volume = {50},
number = {3},
pages = {145},
pmid = {40240583},
issn = {1573-6903},
mesh = {*alpha-Synuclein/metabolism ; *tau Proteins/metabolism ; Humans ; *Parkinson Disease/metabolism/pathology ; *Mitochondria/metabolism/pathology ; *Amyloid beta-Peptides/metabolism ; Animals ; *Nerve Degeneration/metabolism/pathology ; },
abstract = {Parkinson's disease is a complex neurological ailment manifested by dopaminergic neurodegeneration in the substantia nigra of the brain. This study investigates the molecular tripartite interaction between Lewy bodies, amyloid beta, and tau protein in the pathogenesis of Parkinson's disease. Lewy bodies which have been found as the important pathological hallmark in the degenerative neurons of Parkinson's patients, are mainly composed of α-synuclein. The accumulation of α-synuclein has been directly and indirectly linked to the severity and degree of progression of the disease. In addition, approximately 50% of Parkinson's disease cases are also described by hyperphosphorylation of tau protein indicating its significant involvement in the disease. The study further explains how α-synuclein, tau and amyloid beta can spread via cross-seeding mechanisms and accelerate each other's aggregation leading to neuronal death. Both GSK-3β and CDK5 are involved in phosphorylation which among other effects contributes to the misfolding of both α-synuclein and tau proteins that lead to neurodegeneration in Alzheimer's disease. Several mediators, that contribute to mitochondrial damage through elevated oxidative stress pathology are clearly described. Because of the increase in the incidence of Parkinson's disease, as predicted to be 17 million when the study was being conducted, studying these pathological mechanisms is very important in trying to establish treatments. This work contributes a path to finding a multi-target treatment regimen to alleviate the burden of this devastating disease.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*alpha-Synuclein/metabolism
*tau Proteins/metabolism
Humans
*Parkinson Disease/metabolism/pathology
*Mitochondria/metabolism/pathology
*Amyloid beta-Peptides/metabolism
Animals
*Nerve Degeneration/metabolism/pathology
RevDate: 2025-04-16
CmpDate: 2025-04-16
The role of the hippocampus and SLC39A8 in chronic musculoskeletal pain-induced dementia: a Mendelian randomization study.
Scientific reports, 15(1):13211.
Despite observational studies suggesting a link between chronic musculoskeletal pain (CMP) and increased risk of cognitive decline and dementia, the causal nature of this relationship remains uncertain due to potential confounding factors and reverse causality. We employed two-sample Mendelian Randomization (TSMR), bidirectional MR, mediation MR, drug-target MR, and colocalization analysis, along with gene set enrichment and protein-protein interaction (PPI) analyses. TSMR assessed the causal associations between CMP and the risk of dementia and its subtypes, including Alzheimer's disease (AD), vascular dementia (VaD), Lewy body dementia (LBD), frontotemporal dementia (FTD), and Parkinson's disease (PD). Bidirectional MR evaluated reverse causality, while mediation analyses identified potential mediators, focusing on neuroimaging and cognitive phenotypes. Drug-target MR investigated the role of the SLC39A8 gene, and colocalization analysis determined shared causal genetic variants. Gene set enrichment and PPI analyses elucidated the biological pathways implicated in the CMP-dementia relationship. Robust evidence established a causal relationship between chronic low back pain (LBP) and increased risk of PD, with knee osteoarthritis identified as a partial mediator, suggesting a pathway involving chronic inflammation. Bidirectional MR analysis revealed no evidence of reverse causality, further supporting the unidirectional causal link from LBP to PD. Colocalization analysis confirmed distinct genetic architectures for LBP and PD, while drug-target MR implicated the SLC39A8 gene as a potential mediator. Gene set enrichment and PPI analyses highlighted critical biological pathways, such as purine metabolism and glutamate receptor signaling. Suggestive evidence indicated potential causal links between limb pain and overall dementia, myalgia and VaD, as well as potential protective effects of Polymyalgia Rheumatica (PMR) against AD and rheumatism against PD. This study reveals a complex causal relationship between CMP and neurodegenerative diseases, particularly the robust link between LBP and PD. The findings underscore the need for further research to elucidate the underlying mechanisms and inform targeted prevention and treatment strategies.
Additional Links: PMID-40240578
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40240578,
year = {2025},
author = {Du, K and Zuo, YL and Zhang, ZM and Li, A and Zuo, QH and Zhang, CY and Guo, R and Ping, C and Du, WS and Li, SM},
title = {The role of the hippocampus and SLC39A8 in chronic musculoskeletal pain-induced dementia: a Mendelian randomization study.},
journal = {Scientific reports},
volume = {15},
number = {1},
pages = {13211},
pmid = {40240578},
issn = {2045-2322},
mesh = {Humans ; Mendelian Randomization Analysis ; *Dementia/etiology/genetics ; *Hippocampus/metabolism ; *Musculoskeletal Pain/complications/genetics ; *Chronic Pain/complications/genetics ; *Cation Transport Proteins/genetics/metabolism ; Polymorphism, Single Nucleotide ; },
abstract = {Despite observational studies suggesting a link between chronic musculoskeletal pain (CMP) and increased risk of cognitive decline and dementia, the causal nature of this relationship remains uncertain due to potential confounding factors and reverse causality. We employed two-sample Mendelian Randomization (TSMR), bidirectional MR, mediation MR, drug-target MR, and colocalization analysis, along with gene set enrichment and protein-protein interaction (PPI) analyses. TSMR assessed the causal associations between CMP and the risk of dementia and its subtypes, including Alzheimer's disease (AD), vascular dementia (VaD), Lewy body dementia (LBD), frontotemporal dementia (FTD), and Parkinson's disease (PD). Bidirectional MR evaluated reverse causality, while mediation analyses identified potential mediators, focusing on neuroimaging and cognitive phenotypes. Drug-target MR investigated the role of the SLC39A8 gene, and colocalization analysis determined shared causal genetic variants. Gene set enrichment and PPI analyses elucidated the biological pathways implicated in the CMP-dementia relationship. Robust evidence established a causal relationship between chronic low back pain (LBP) and increased risk of PD, with knee osteoarthritis identified as a partial mediator, suggesting a pathway involving chronic inflammation. Bidirectional MR analysis revealed no evidence of reverse causality, further supporting the unidirectional causal link from LBP to PD. Colocalization analysis confirmed distinct genetic architectures for LBP and PD, while drug-target MR implicated the SLC39A8 gene as a potential mediator. Gene set enrichment and PPI analyses highlighted critical biological pathways, such as purine metabolism and glutamate receptor signaling. Suggestive evidence indicated potential causal links between limb pain and overall dementia, myalgia and VaD, as well as potential protective effects of Polymyalgia Rheumatica (PMR) against AD and rheumatism against PD. This study reveals a complex causal relationship between CMP and neurodegenerative diseases, particularly the robust link between LBP and PD. The findings underscore the need for further research to elucidate the underlying mechanisms and inform targeted prevention and treatment strategies.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Mendelian Randomization Analysis
*Dementia/etiology/genetics
*Hippocampus/metabolism
*Musculoskeletal Pain/complications/genetics
*Chronic Pain/complications/genetics
*Cation Transport Proteins/genetics/metabolism
Polymorphism, Single Nucleotide
RevDate: 2025-04-16
Activation of Epac2 improves Aβ-induced impairment of memory retrieval in an acute model of Alzheimer's disease.
Neuropharmacology pii:S0028-3908(25)00174-1 [Epub ahead of print].
Impaired memory retrieval is one of the cognitive markers in the early stage of Alzheimer's Disease (AD). Previous studies report that exchange protein directly activated by cAMP 2 (Epac2) plays a specific and time-limited role in promoting memory retrieval. In this study, we investigated the effect of a novel Epac2 activator, S220, on neuronal and synaptic activities, and memory impairment in an acute AD mouse model. S220 treatment increased the firing rate of action potential and intracellular calcium in primary neurons. Moreover, S220 treatment increased synaptic currents in CA1 neurons. In the acute AD mouse model, intrahippocampal injection of amyloid-β (Aβ) oligomers impaired memory performance. Notably, administering S220 20 minutes before retention of contextual fear conditioning recovered the Aβ-induced memory impairment, suggesting an enhancing effect on memory retrieval. Collectively, our data demonstrate that the novel Epac2 activator S220 promotes synaptic communication and neuronal firing, and thereby improves Aβ-induced memory impairment via enhancing memory retrieval, indicating the role of Epac2 as a potential treatment target for AD.
Additional Links: PMID-40239917
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40239917,
year = {2025},
author = {Zhang, T and Zhang, Y and Chameau, P and Chen, T and Marmolejo-Garza, A and Douwenga, W and Dolga, AM and Kessels, HW and Schmidt, M and Eisel, ULM},
title = {Activation of Epac2 improves Aβ-induced impairment of memory retrieval in an acute model of Alzheimer's disease.},
journal = {Neuropharmacology},
volume = {},
number = {},
pages = {110468},
doi = {10.1016/j.neuropharm.2025.110468},
pmid = {40239917},
issn = {1873-7064},
abstract = {Impaired memory retrieval is one of the cognitive markers in the early stage of Alzheimer's Disease (AD). Previous studies report that exchange protein directly activated by cAMP 2 (Epac2) plays a specific and time-limited role in promoting memory retrieval. In this study, we investigated the effect of a novel Epac2 activator, S220, on neuronal and synaptic activities, and memory impairment in an acute AD mouse model. S220 treatment increased the firing rate of action potential and intracellular calcium in primary neurons. Moreover, S220 treatment increased synaptic currents in CA1 neurons. In the acute AD mouse model, intrahippocampal injection of amyloid-β (Aβ) oligomers impaired memory performance. Notably, administering S220 20 minutes before retention of contextual fear conditioning recovered the Aβ-induced memory impairment, suggesting an enhancing effect on memory retrieval. Collectively, our data demonstrate that the novel Epac2 activator S220 promotes synaptic communication and neuronal firing, and thereby improves Aβ-induced memory impairment via enhancing memory retrieval, indicating the role of Epac2 as a potential treatment target for AD.},
}
RevDate: 2025-04-16
Neuropharmacological effects of calycosin: a translational review of molecular mechanisms and therapeutic applications.
Naunyn-Schmiedeberg's archives of pharmacology [Epub ahead of print].
Calycosin, a naturally occurring isoflavonoid found predominantly in Astragalus membranaceus, exhibits significant therapeutic potential in various neurological conditions. Its multifaceted bioactive properties-antioxidant, anti-inflammatory, and anti-apoptotic-position it as a promising candidate for neuroprotection and neuroregeneration. This review explores calycosin's mechanisms of action, including its modulation of key signaling pathways such as HMGB1/TLR4/NF-κB (high mobility group box 1/toll-like receptor 4/nuclear factor kappa B), phosphatidylinositol-3-kinase (PI3 K)/Akt, ERK1/2 (extracellular signal-regulated kinase 1/2), and Hsp90/Akt/p38. In cerebral ischemia/reperfusion injury, calycosin reduces oxidative stress markers like ROS (reactive oxygen species) and MDA (malondialdehyde), enhances antioxidant enzymes (SOD (superoxide dismutase) and GPX (glutathione peroxidase)), and downregulates pro-inflammatory cytokines (TNF-α, IL-1β) through the HMGB1/TLR4/NF-κB pathway. It also inhibits autophagy via the STAT3/FOXO3a pathway and apoptosis by modulating Bax and Bcl-2 expression. In neuro-oncology, calycosin inhibits glioblastoma cell migration and invasion by modulating the TGF-β-mediated mesenchymal properties and suppressing the c-Met and CXCL10 signaling pathways. Additionally, it enhances the efficacy of temozolomide in glioma treatment through apoptotic pathways involving caspase-3 and caspase-9. Calycosin shows promise in Alzheimer's disease by reducing β-amyloid production and tau hyperphosphorylation via the GSK-3β pathway and improving mitochondrial function through the peroxisome proliferator-activated receptor gamma coactivator 1-Alpha (PGC-1α)/mitochondrial transcription factor A (TFAM) signaling pathway. In Parkinson's disease, calycosin mitigates oxidative stress, prevents dopaminergic neuronal death, and reduces neuroinflammation by inhibiting the TLR/NF-κB and MAPK pathways. It has also shown therapeutic potential in meningitis and even neuroprotective effects against hyperbilirubinemia-induced nerve injury. Despite these promising findings, further research, including detailed mechanistic studies and clinical trials, is needed to fully understand calycosin's therapeutic mechanisms and validate its potential in human subjects. Developing advanced delivery systems and exploring synergistic therapeutic strategies could further enhance its clinical application and effectiveness.
Additional Links: PMID-40237798
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40237798,
year = {2025},
author = {Ahmed, HS},
title = {Neuropharmacological effects of calycosin: a translational review of molecular mechanisms and therapeutic applications.},
journal = {Naunyn-Schmiedeberg's archives of pharmacology},
volume = {},
number = {},
pages = {},
pmid = {40237798},
issn = {1432-1912},
abstract = {Calycosin, a naturally occurring isoflavonoid found predominantly in Astragalus membranaceus, exhibits significant therapeutic potential in various neurological conditions. Its multifaceted bioactive properties-antioxidant, anti-inflammatory, and anti-apoptotic-position it as a promising candidate for neuroprotection and neuroregeneration. This review explores calycosin's mechanisms of action, including its modulation of key signaling pathways such as HMGB1/TLR4/NF-κB (high mobility group box 1/toll-like receptor 4/nuclear factor kappa B), phosphatidylinositol-3-kinase (PI3 K)/Akt, ERK1/2 (extracellular signal-regulated kinase 1/2), and Hsp90/Akt/p38. In cerebral ischemia/reperfusion injury, calycosin reduces oxidative stress markers like ROS (reactive oxygen species) and MDA (malondialdehyde), enhances antioxidant enzymes (SOD (superoxide dismutase) and GPX (glutathione peroxidase)), and downregulates pro-inflammatory cytokines (TNF-α, IL-1β) through the HMGB1/TLR4/NF-κB pathway. It also inhibits autophagy via the STAT3/FOXO3a pathway and apoptosis by modulating Bax and Bcl-2 expression. In neuro-oncology, calycosin inhibits glioblastoma cell migration and invasion by modulating the TGF-β-mediated mesenchymal properties and suppressing the c-Met and CXCL10 signaling pathways. Additionally, it enhances the efficacy of temozolomide in glioma treatment through apoptotic pathways involving caspase-3 and caspase-9. Calycosin shows promise in Alzheimer's disease by reducing β-amyloid production and tau hyperphosphorylation via the GSK-3β pathway and improving mitochondrial function through the peroxisome proliferator-activated receptor gamma coactivator 1-Alpha (PGC-1α)/mitochondrial transcription factor A (TFAM) signaling pathway. In Parkinson's disease, calycosin mitigates oxidative stress, prevents dopaminergic neuronal death, and reduces neuroinflammation by inhibiting the TLR/NF-κB and MAPK pathways. It has also shown therapeutic potential in meningitis and even neuroprotective effects against hyperbilirubinemia-induced nerve injury. Despite these promising findings, further research, including detailed mechanistic studies and clinical trials, is needed to fully understand calycosin's therapeutic mechanisms and validate its potential in human subjects. Developing advanced delivery systems and exploring synergistic therapeutic strategies could further enhance its clinical application and effectiveness.},
}
RevDate: 2025-04-16
Intervention points for the role of physical activity in prevention and treatment of Alzheimer's disease.
The Journal of physiology [Epub ahead of print].
Alzheimer's disease (AD) is a growing global health challenge with limited pharmacological treatments. Epidemiological studies link regular physical activity with a lower risk of AD and cognitive decline in general, whereas randomized controlled trials show that aerobic exercise slows disease progression and improves cognitive function. However the underlying mechanisms remain incompletely understood. In this review we discuss five likely intervention points through which physical activity may influence AD progression and pathology: (1) reducing neuroinflammation and amyloid beta (Aβ) aggregation, (2) enhancing clearance of Aβ aggregates, (3) increasing neuronal resilience, (4) promoting hippocampal neurogenesis and (5) strengthening cognitive reserve. Understanding which of these mechanistic links are most likely to drive the AD-protective effects of exercise could help refine lifestyle-based interventions to complement pharmacological treatments and inform future prevention strategies.
Additional Links: PMID-40237393
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40237393,
year = {2025},
author = {Brendborg, N and Febbraio, MA},
title = {Intervention points for the role of physical activity in prevention and treatment of Alzheimer's disease.},
journal = {The Journal of physiology},
volume = {},
number = {},
pages = {},
doi = {10.1113/JP286747},
pmid = {40237393},
issn = {1469-7793},
support = {1194141//DHAC | National Health and Medical Research Council (NHMRC)/ ; },
abstract = {Alzheimer's disease (AD) is a growing global health challenge with limited pharmacological treatments. Epidemiological studies link regular physical activity with a lower risk of AD and cognitive decline in general, whereas randomized controlled trials show that aerobic exercise slows disease progression and improves cognitive function. However the underlying mechanisms remain incompletely understood. In this review we discuss five likely intervention points through which physical activity may influence AD progression and pathology: (1) reducing neuroinflammation and amyloid beta (Aβ) aggregation, (2) enhancing clearance of Aβ aggregates, (3) increasing neuronal resilience, (4) promoting hippocampal neurogenesis and (5) strengthening cognitive reserve. Understanding which of these mechanistic links are most likely to drive the AD-protective effects of exercise could help refine lifestyle-based interventions to complement pharmacological treatments and inform future prevention strategies.},
}
RevDate: 2025-04-16
CmpDate: 2025-04-16
Whispers in the Brain: Extracellular Vesicles in Neuropathology and the Diagnostic Alchemy of Neurological Diseases.
The European journal of neuroscience, 61(8):e70090.
Extracellular vesicles (EVs) have emerged as pivotal mediators in neurological diseases, showcasing multifaceted potential roles ranging from pathogenesis to diagnosis. These nano-sized membranous structures, released by various cell types including neurons, astrocytes, and microglia, encapsulate a diverse cargo of proteins, lipids, RNA species, and even DNA fragments. In neuropathology, EVs contribute significantly to intercellular communication within the central nervous system (CNS), influencing physiological or pathological cascades. Through the transfer of bioactive molecules, EVs modulate neuroinflammation, neuronal survival, synaptic plasticity, and the propagation of protein aggregates characteristic of neurodegenerative disorders. Moreover, their presence in biofluids such as cerebrospinal fluid (CSF), blood, and urine reflects the pathophysiological state of the CNS, offering a window into the diagnosis, monitoring and treatment of neurological diseases. Recent advancements in EV isolation techniques, coupled with high-throughput omics technologies, have facilitated the profiling of EV cargo, enabling the identification of disease-specific biomarkers with high sensitivity and specificity. This review explores the intricate roles of EVs in neuropathology, highlighting their involvement in Alzheimer's disease, Parkinson's disease, multiple sclerosis, and other neurological disorders. Furthermore, it delves into the diagnostic potential of EVs, discussing current challenges and prospects in harnessing EV-derived biomarkers for precision medicine in neurology. Ultimately, understanding the biology of EVs in neurological contexts promises transformative insights into disease mechanisms and therapeutic strategies, paving the way for innovative diagnostic tools and targeted interventions in clinical practice.
Additional Links: PMID-40237381
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40237381,
year = {2025},
author = {Isik, M and Sari, HK and Caglayan, MG and Yilmaz, R and Derkus, B},
title = {Whispers in the Brain: Extracellular Vesicles in Neuropathology and the Diagnostic Alchemy of Neurological Diseases.},
journal = {The European journal of neuroscience},
volume = {61},
number = {8},
pages = {e70090},
doi = {10.1111/ejn.70090},
pmid = {40237381},
issn = {1460-9568},
support = {TSA-2023-2648//Ankara University Scientific Research Projects Coordination Unit/ ; TSA-2024-3217//Ankara University Scientific Research Projects Coordination Unit/ ; },
mesh = {Humans ; *Extracellular Vesicles/metabolism/pathology ; Animals ; *Nervous System Diseases/diagnosis/metabolism/pathology ; Biomarkers/metabolism ; *Brain/metabolism/pathology ; Neurodegenerative Diseases/diagnosis/metabolism ; },
abstract = {Extracellular vesicles (EVs) have emerged as pivotal mediators in neurological diseases, showcasing multifaceted potential roles ranging from pathogenesis to diagnosis. These nano-sized membranous structures, released by various cell types including neurons, astrocytes, and microglia, encapsulate a diverse cargo of proteins, lipids, RNA species, and even DNA fragments. In neuropathology, EVs contribute significantly to intercellular communication within the central nervous system (CNS), influencing physiological or pathological cascades. Through the transfer of bioactive molecules, EVs modulate neuroinflammation, neuronal survival, synaptic plasticity, and the propagation of protein aggregates characteristic of neurodegenerative disorders. Moreover, their presence in biofluids such as cerebrospinal fluid (CSF), blood, and urine reflects the pathophysiological state of the CNS, offering a window into the diagnosis, monitoring and treatment of neurological diseases. Recent advancements in EV isolation techniques, coupled with high-throughput omics technologies, have facilitated the profiling of EV cargo, enabling the identification of disease-specific biomarkers with high sensitivity and specificity. This review explores the intricate roles of EVs in neuropathology, highlighting their involvement in Alzheimer's disease, Parkinson's disease, multiple sclerosis, and other neurological disorders. Furthermore, it delves into the diagnostic potential of EVs, discussing current challenges and prospects in harnessing EV-derived biomarkers for precision medicine in neurology. Ultimately, understanding the biology of EVs in neurological contexts promises transformative insights into disease mechanisms and therapeutic strategies, paving the way for innovative diagnostic tools and targeted interventions in clinical practice.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Extracellular Vesicles/metabolism/pathology
Animals
*Nervous System Diseases/diagnosis/metabolism/pathology
Biomarkers/metabolism
*Brain/metabolism/pathology
Neurodegenerative Diseases/diagnosis/metabolism
RevDate: 2025-04-16
CmpDate: 2025-04-16
Lecanemab preferentially binds to smaller aggregates present at early Alzheimer's disease.
Alzheimer's & dementia : the journal of the Alzheimer's Association, 21(4):e70086.
INTRODUCTION: The monoclonal antibodies Aducanumab, Lecanemab, Gantenerumab, and Donanemab were developed for the treatment of Alzheimer's disease (AD).
METHODS: We used single-molecule detection and super-resolution imaging to characterize the binding of these antibodies to diffusible amyloid beta (Aβ) aggregates generated in-vitro and harvested from human brains.
RESULTS: Lecanemab showed the best performance in terms of binding to the small-diffusible Aβ aggregates, affinity, aggregate coating, and the ability to bind to post-translationally modified species, providing an explanation for its therapeutic success. We observed a Braak stage-dependent increase in small-diffusible aggregate quantity and size, which was detectable with Aducanumab and Gantenerumab, but not Lecanemab, showing that the diffusible Aβ aggregates change with disease progression and the smaller aggregates to which Lecanemab preferably binds exist at higher quantities during earlier stages.
DISCUSSION: These findings provide an explanation for the success of Lecanemab in clinical trials and suggests that Lecanemab will be more effective when used in early-stage AD.
HIGHLIGHTS: Anti amyloid beta therapeutics are compared by their diffusible aggregate binding characteristics. In-vitro and brain-derived aggregates are tested using single-molecule detection. Lecanemab shows therapeutic success by binding to aggregates formed in early disease. Lecanemab binds to these aggregates with high affinity and coats them better.
Additional Links: PMID-40237235
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40237235,
year = {2025},
author = {Fertan, E and Lam, JYL and Albertini, G and Dewilde, M and Wu, Y and Akingbade, OES and Böken, D and English, EA and De Strooper, B and Klenerman, D},
title = {Lecanemab preferentially binds to smaller aggregates present at early Alzheimer's disease.},
journal = {Alzheimer's & dementia : the journal of the Alzheimer's Association},
volume = {21},
number = {4},
pages = {e70086},
doi = {10.1002/alz.70086},
pmid = {40237235},
issn = {1552-5279},
support = {ERC-834682//European Union's Horizon 2020 Research and Innovation Program/ ; AARF-22-968623//Alzheimer's Association USA/ ; MR/Y014847/1//UK Medical Research Council/ ; //UK Dementia Research Institute/ ; //The Royal Society/ ; },
mesh = {*Alzheimer Disease/metabolism/drug therapy/pathology ; Humans ; *Amyloid beta-Peptides/metabolism ; *Antibodies, Monoclonal, Humanized/metabolism/pharmacology ; *Brain/metabolism/pathology ; *Antibodies, Monoclonal/metabolism ; },
abstract = {INTRODUCTION: The monoclonal antibodies Aducanumab, Lecanemab, Gantenerumab, and Donanemab were developed for the treatment of Alzheimer's disease (AD).
METHODS: We used single-molecule detection and super-resolution imaging to characterize the binding of these antibodies to diffusible amyloid beta (Aβ) aggregates generated in-vitro and harvested from human brains.
RESULTS: Lecanemab showed the best performance in terms of binding to the small-diffusible Aβ aggregates, affinity, aggregate coating, and the ability to bind to post-translationally modified species, providing an explanation for its therapeutic success. We observed a Braak stage-dependent increase in small-diffusible aggregate quantity and size, which was detectable with Aducanumab and Gantenerumab, but not Lecanemab, showing that the diffusible Aβ aggregates change with disease progression and the smaller aggregates to which Lecanemab preferably binds exist at higher quantities during earlier stages.
DISCUSSION: These findings provide an explanation for the success of Lecanemab in clinical trials and suggests that Lecanemab will be more effective when used in early-stage AD.
HIGHLIGHTS: Anti amyloid beta therapeutics are compared by their diffusible aggregate binding characteristics. In-vitro and brain-derived aggregates are tested using single-molecule detection. Lecanemab shows therapeutic success by binding to aggregates formed in early disease. Lecanemab binds to these aggregates with high affinity and coats them better.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/metabolism/drug therapy/pathology
Humans
*Amyloid beta-Peptides/metabolism
*Antibodies, Monoclonal, Humanized/metabolism/pharmacology
*Brain/metabolism/pathology
*Antibodies, Monoclonal/metabolism
RevDate: 2025-04-16
Photodynamic and Photothermal Therapies using Nanotechnology Approach in Alzheimer's Disease.
Current neuropharmacology pii:CN-EPUB-147677 [Epub ahead of print].
Alzheimer's disease is a neurodegenerative disease that impairs cognitive function. The incidence of Alzheimer's disease increases with the increase in the elderly population. Although the clear pathogenesis of Alzheimer's disease is not yet known, the formation of amyloid plaques and tau fibrils, diminished acetylcholine levels, and increased inflammation can be observed in patients. Alzheimer's disease, whose pathogenesis is not fully demonstrated, cannot be treated radically. Since it has been observed that only pharmacological treatment alone isn't sufficient, alternative approaches have become essential. Among these approaches, nanocarriers greatly facilitate the transport of drugs since the blood-brain barrier is an important obstacle to the penetration of drugs into the brain. Photosensitizers trigger activation after exposure to near-infrared radiation light of a suitable wavelength or laser light, resulting in the selective destruction of Aβ plaques. Photodynamic therapy and photothermal therapy have been investigated for their potential to inhibit Aβ plaques through photosensitizers. By encapsulating photosensitizers in nanocarriers, the limitations of photosensitizers can be overcome. By using these photosensitizers, near-infrared radiation fluorescence imaging can be used as a theranostic. In this review, potential treatment options for photodynamic therapy and photothermal therapy for Alzheimer's disease are summarised, and a simultaneous or combined approach is discussed, taking into account potential nanotheranostics.
Additional Links: PMID-40237061
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40237061,
year = {2025},
author = {Öztürk, B and Demir, H and Günay, MS and Akdag, Y and Sahin, S and Gulsun, T},
title = {Photodynamic and Photothermal Therapies using Nanotechnology Approach in Alzheimer's Disease.},
journal = {Current neuropharmacology},
volume = {},
number = {},
pages = {},
doi = {10.2174/011570159X370790250317045223},
pmid = {40237061},
issn = {1875-6190},
abstract = {Alzheimer's disease is a neurodegenerative disease that impairs cognitive function. The incidence of Alzheimer's disease increases with the increase in the elderly population. Although the clear pathogenesis of Alzheimer's disease is not yet known, the formation of amyloid plaques and tau fibrils, diminished acetylcholine levels, and increased inflammation can be observed in patients. Alzheimer's disease, whose pathogenesis is not fully demonstrated, cannot be treated radically. Since it has been observed that only pharmacological treatment alone isn't sufficient, alternative approaches have become essential. Among these approaches, nanocarriers greatly facilitate the transport of drugs since the blood-brain barrier is an important obstacle to the penetration of drugs into the brain. Photosensitizers trigger activation after exposure to near-infrared radiation light of a suitable wavelength or laser light, resulting in the selective destruction of Aβ plaques. Photodynamic therapy and photothermal therapy have been investigated for their potential to inhibit Aβ plaques through photosensitizers. By encapsulating photosensitizers in nanocarriers, the limitations of photosensitizers can be overcome. By using these photosensitizers, near-infrared radiation fluorescence imaging can be used as a theranostic. In this review, potential treatment options for photodynamic therapy and photothermal therapy for Alzheimer's disease are summarised, and a simultaneous or combined approach is discussed, taking into account potential nanotheranostics.},
}
RevDate: 2025-04-16
Homeostatic Activation of 26S Proteasomes by Protein Kinase A Protects against Cardiac and Neurobehavior Malfunction in Alzheimer's Disease Mice.
bioRxiv : the preprint server for biology pii:2025.03.28.645869.
Alzheimer's Disease (AD) patients often show brain and cardiac malfunction. AD represents a leading cause of morbidity and mortality worldwide, but the demand for effective treatment for AD is far from being met. This is primarily because AD pathogenesis, including brain-heart interaction, is poorly understood. Proteasome functional insufficiency is implicated in AD; as such, proteasome enhancement promises a potentially new strategy to treat AD. The proteasome can be activated by protein kinase A (PKA) via selectively phosphorylating Ser14-RPN6/PSMD11 (p-S14-RPN6); however, whether p-S14-RPN6 is altered and what role p-S14-RPN6 plays in AD remain unclear. Hence, this study was conducted to address these critical gaps. We found that genetic blockade of the homeostatic p-S14-Rpn6 via germline knock-in of Rpn6 [S14A] (referred to as S14A) significantly reduced proteasome activities in the cerebral cortex but did not discernibly impair learning and memory function in 4-month-old mice or cause cardiac dysfunction before 12 months of age. Increases in Ser14-phosphorylated Rpn6 in the cerebral cortex and markedly elevated Aβ proteins in the myocardium were observed in young 5XFAD mice, a commonly used AD model. When introduced into the 5XFAD mice, S14A significantly aggravated the learning and memory deficits as revealed by the radial arm water maze tests and accelerated cardiac malfunction as measured by serial echocardiography in the same cohort of 5XFAD mice. Thus, the present study establishes for the first time that homeostatic activation of 26S proteasomes by basal p-S14-RPN6 or PKA activity protects against both the brain and heart malfunction in the 5XFAD mice.
Additional Links: PMID-40236239
Full Text:
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40236239,
year = {2025},
author = {Chakroborty, A and Ejaz, S and Sternburg, JO and Asadi, Y and Cai, M and Dwamena, AA and Giri, S and Adeniji, O and Ahammed, MS and Gilstrap, EA and Uddin, MG and McDowell, C and Liu, J and Wang, H and Wang, X},
title = {Homeostatic Activation of 26S Proteasomes by Protein Kinase A Protects against Cardiac and Neurobehavior Malfunction in Alzheimer's Disease Mice.},
journal = {bioRxiv : the preprint server for biology},
volume = {},
number = {},
pages = {},
doi = {10.1101/2025.03.28.645869},
pmid = {40236239},
issn = {2692-8205},
abstract = {Alzheimer's Disease (AD) patients often show brain and cardiac malfunction. AD represents a leading cause of morbidity and mortality worldwide, but the demand for effective treatment for AD is far from being met. This is primarily because AD pathogenesis, including brain-heart interaction, is poorly understood. Proteasome functional insufficiency is implicated in AD; as such, proteasome enhancement promises a potentially new strategy to treat AD. The proteasome can be activated by protein kinase A (PKA) via selectively phosphorylating Ser14-RPN6/PSMD11 (p-S14-RPN6); however, whether p-S14-RPN6 is altered and what role p-S14-RPN6 plays in AD remain unclear. Hence, this study was conducted to address these critical gaps. We found that genetic blockade of the homeostatic p-S14-Rpn6 via germline knock-in of Rpn6 [S14A] (referred to as S14A) significantly reduced proteasome activities in the cerebral cortex but did not discernibly impair learning and memory function in 4-month-old mice or cause cardiac dysfunction before 12 months of age. Increases in Ser14-phosphorylated Rpn6 in the cerebral cortex and markedly elevated Aβ proteins in the myocardium were observed in young 5XFAD mice, a commonly used AD model. When introduced into the 5XFAD mice, S14A significantly aggravated the learning and memory deficits as revealed by the radial arm water maze tests and accelerated cardiac malfunction as measured by serial echocardiography in the same cohort of 5XFAD mice. Thus, the present study establishes for the first time that homeostatic activation of 26S proteasomes by basal p-S14-RPN6 or PKA activity protects against both the brain and heart malfunction in the 5XFAD mice.},
}
RevDate: 2025-04-16
Cognitive Deficits in Late-Life Depression: From Symptoms and Assessment to Therapeutics.
Focus (American Psychiatric Publishing), 23(2):183-194.
Cognitive symptoms and deficits are core features of late-life depression (LLD), with an estimated 20%-50% of affected individuals meeting diagnostic criteria for mild cognitive impairment (MCI). Cognitive deficits, especially executive dysfunction, have consistently been associated with poorer treatment outcomes among people with LLD. Furthermore, distinguishing depression with cognitive complaints or cognitive impairment from the early stages of Alzheimer's disease (AD) can be challenging. Cognitive concerns are often emphasized among those with LLD, although, paradoxically, their description of memory difficulty may include detailed recall of specific memory lapses. Conversely, people with AD often have limited insight into their progressive cognitive decline, minimizing and concealing their cognitive difficulties. Neuropsychological assessment is one of the most useful means of clarifying this differential diagnosis. A subcortical cognitive pattern is commonly observed among people with LLD, including psychomotor slowing, variable attention, and executive dysfunction, which can affect memory encoding and free recall. A broad range of therapeutic approaches have been applied to older adults experiencing LLD along with cognitive symptoms, MCI, or dementia. Most studies focus on treatments to address LLD or MCI, with relatively fewer examining treatments specifically at this intersection. Nonpharmacological strategies, including aerobic exercise, cognitive remediation, and neuromodulation, are highly recommended to improve both depression and cognition. Antidepressants may have benefits for elements of cognition among people with LLD, but they have less evidence for their efficacy for people with cognitive deficits and dementia. This review provides an updated conceptual and practical framework for clinicians evaluating and treating LLD.
Additional Links: PMID-40235602
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40235602,
year = {2025},
author = {Teixeira, AL and Gregg, A and Gentry, MT and Gujral, S and Rapp, E and Oberlin, L and Ajilore, O and Weisenbach, S and Patrick, R},
title = {Cognitive Deficits in Late-Life Depression: From Symptoms and Assessment to Therapeutics.},
journal = {Focus (American Psychiatric Publishing)},
volume = {23},
number = {2},
pages = {183-194},
pmid = {40235602},
issn = {1541-4094},
abstract = {Cognitive symptoms and deficits are core features of late-life depression (LLD), with an estimated 20%-50% of affected individuals meeting diagnostic criteria for mild cognitive impairment (MCI). Cognitive deficits, especially executive dysfunction, have consistently been associated with poorer treatment outcomes among people with LLD. Furthermore, distinguishing depression with cognitive complaints or cognitive impairment from the early stages of Alzheimer's disease (AD) can be challenging. Cognitive concerns are often emphasized among those with LLD, although, paradoxically, their description of memory difficulty may include detailed recall of specific memory lapses. Conversely, people with AD often have limited insight into their progressive cognitive decline, minimizing and concealing their cognitive difficulties. Neuropsychological assessment is one of the most useful means of clarifying this differential diagnosis. A subcortical cognitive pattern is commonly observed among people with LLD, including psychomotor slowing, variable attention, and executive dysfunction, which can affect memory encoding and free recall. A broad range of therapeutic approaches have been applied to older adults experiencing LLD along with cognitive symptoms, MCI, or dementia. Most studies focus on treatments to address LLD or MCI, with relatively fewer examining treatments specifically at this intersection. Nonpharmacological strategies, including aerobic exercise, cognitive remediation, and neuromodulation, are highly recommended to improve both depression and cognition. Antidepressants may have benefits for elements of cognition among people with LLD, but they have less evidence for their efficacy for people with cognitive deficits and dementia. This review provides an updated conceptual and practical framework for clinicians evaluating and treating LLD.},
}
RevDate: 2025-04-16
CmpDate: 2025-04-16
Circular RNAs, miRNAs, and Exosomes: Their Roles and Importance in Amyloid-Beta and Tau Pathologies in Alzheimer's Disease.
Neural plasticity, 2025:9581369.
Alzheimer's disease (AD) is a devastating neurodegenerative disorder. The pathology of this disease is based on two basic mechanisms: amyloid-beta (Aβ) and tau fibrillation. Many genes and mechanisms have been identified as the primary causes of AD in clinical settings, and there have been exciting developments in drug treatments. Several molecules and biological structures regulate the genome outside of the standard DNA function. As in many diseases, circular RNAs (circRNAs), microRNAs (miRNAs), and exosomes (EXOs), investigated from different aspects of AD, are useful for treatment and diagnosis. This review examines two biological elements regarding their roles in the Aβ-tau pathology of AD and their potential as treatment targets. Importantly, the activities of miRNAs that play a role in these processes were evaluated. Trial Registration: ClinicalTrials.gov identifiers: NCT04120493, NCT04969172, NCT04388982.
Additional Links: PMID-40235521
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40235521,
year = {2025},
author = {Pala, M and Yilmaz, SG},
title = {Circular RNAs, miRNAs, and Exosomes: Their Roles and Importance in Amyloid-Beta and Tau Pathologies in Alzheimer's Disease.},
journal = {Neural plasticity},
volume = {2025},
number = {},
pages = {9581369},
pmid = {40235521},
issn = {1687-5443},
mesh = {*Alzheimer Disease/metabolism/pathology/genetics ; Humans ; *RNA, Circular/metabolism ; *Exosomes/metabolism ; *Amyloid beta-Peptides/metabolism ; *MicroRNAs/metabolism ; *tau Proteins/metabolism ; Animals ; },
abstract = {Alzheimer's disease (AD) is a devastating neurodegenerative disorder. The pathology of this disease is based on two basic mechanisms: amyloid-beta (Aβ) and tau fibrillation. Many genes and mechanisms have been identified as the primary causes of AD in clinical settings, and there have been exciting developments in drug treatments. Several molecules and biological structures regulate the genome outside of the standard DNA function. As in many diseases, circular RNAs (circRNAs), microRNAs (miRNAs), and exosomes (EXOs), investigated from different aspects of AD, are useful for treatment and diagnosis. This review examines two biological elements regarding their roles in the Aβ-tau pathology of AD and their potential as treatment targets. Importantly, the activities of miRNAs that play a role in these processes were evaluated. Trial Registration: ClinicalTrials.gov identifiers: NCT04120493, NCT04969172, NCT04388982.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Alzheimer Disease/metabolism/pathology/genetics
Humans
*RNA, Circular/metabolism
*Exosomes/metabolism
*Amyloid beta-Peptides/metabolism
*MicroRNAs/metabolism
*tau Proteins/metabolism
Animals
RevDate: 2025-04-16
Data-Driven Modeling of Amyloid-beta Targeted Antibodies for Alzheimer's Disease.
Research square pii:rs.3.rs-6316455.
Alzheimer's disease (AD) is caused by the build-up of amyloid beta (A$\beta$) proteins in the brain, leading to memory loss and cognitive decline. Despite the approval of monoclonal antibodies targeting A$\beta$, optimizing treatment strategies while minimizing side effects remains a challenge. This study develops a mathematical framework to model A$\beta$ aggregation dynamics, capturing the transition from monomers to higher-order aggregates, including protofibrils, toxic oligomers, and fibrils, using mass-action kinetics and coarse-grained modeling. Parameter estimation, sensitivity analysis, and data-driven calibration ensure model robustness. An optimal control framework is introduced to identify the optimal dose of the drug as a control function that reduces toxic oligomers and fibrils while minimizing adverse effects, such as amyloid-related imaging abnormalities (ARIA). The results indicate that Donanemab achieves the most significant reduction in fibrils. These findings provide a quantitative basis for optimizing AD treatments, providing valuable insight into the balance between therapeutic efficacy and safety.
Additional Links: PMID-40235513
Full Text:
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40235513,
year = {2025},
author = {Rabiei, K and Petrella, JR and Lenhart, S and Liu, C and Doraiswamy, PM and Hao, W},
title = {Data-Driven Modeling of Amyloid-beta Targeted Antibodies for Alzheimer's Disease.},
journal = {Research square},
volume = {},
number = {},
pages = {},
doi = {10.21203/rs.3.rs-6316455/v1},
pmid = {40235513},
issn = {2693-5015},
abstract = {Alzheimer's disease (AD) is caused by the build-up of amyloid beta (A$\beta$) proteins in the brain, leading to memory loss and cognitive decline. Despite the approval of monoclonal antibodies targeting A$\beta$, optimizing treatment strategies while minimizing side effects remains a challenge. This study develops a mathematical framework to model A$\beta$ aggregation dynamics, capturing the transition from monomers to higher-order aggregates, including protofibrils, toxic oligomers, and fibrils, using mass-action kinetics and coarse-grained modeling. Parameter estimation, sensitivity analysis, and data-driven calibration ensure model robustness. An optimal control framework is introduced to identify the optimal dose of the drug as a control function that reduces toxic oligomers and fibrils while minimizing adverse effects, such as amyloid-related imaging abnormalities (ARIA). The results indicate that Donanemab achieves the most significant reduction in fibrils. These findings provide a quantitative basis for optimizing AD treatments, providing valuable insight into the balance between therapeutic efficacy and safety.},
}
RevDate: 2025-04-16
Effect of Amyloid Beta on Cholesterol Metabolism-Correlated microRNAs in Primary Cultured Astrocytes of C57BL/6J Mice: A Focus on CYP46A1 and APOE Genes.
Cell journal, 26(11):625-631 pii:720543.
OBJECTIVE: The accumulation of amyloid plaques and disturbance of cholesterol homeostasis are implicated in the pathophysiology of Alzheimer's disease. Apolipoprotein E (ApoE) and cholesterol 24-hydroxylase (CYP46A1) are key proteins involved in the efflux and metabolism of excess cholesterol, and small non-coding RNAs (miRNAs), can help to regulate the expression of the genes encoding these proteins. The aim of the present study was to investigate the alterations in the expression of APOE and CYP46A1 genes, as well as their respective regulatory miRNAs, in astrocytes treated with amyloid beta (Aβ).
MATERIALS AND METHODS: In this experimental study, isolated astrocyte cells were cultured and treated with Aβ for 24 hours. Changes in the expression of APOE and CYP46A1 genes, as well as their regulating miRNAs, were assessed using the realtime polymerase chain reaction (PCR) technique.
RESULTS: The expression of APOE and CYP46A1 genes increased with Aβ treatment. MiR-33a-5p, as the negative regulator of the APOE gene exhibited significant decrease. Additionally, miR-let-7a-5p, as the positive regulator of the APOE gene, showed an increase in the Aβ treated group. Moreover, miR-98-5p, as the negative regulator of the CYP46A1 gene, showed a half-fold decrease. While, miR-27a-3p as the positive regulator of the CYP46A1 gene, increased significantly with Aβ treatment.
CONCLUSION: Alterations in the expression of APOE and CYP46A1 genes, as well as the expression of miRNAs regulating these genes, in astrocytes treated with Aβ suggests that the cell is attempting to modify the regulatory pathways of cholesterol homeostasis in the brain under pathological conditions, such as Alzheimer's disease.
Additional Links: PMID-40235143
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40235143,
year = {2025},
author = {Jaberian Asl, B and Nazeri, Z and Pezeshki, SP and Kheirollah, A and Azizidoost, S and Adelipour, M and Cheraghzadeh, M},
title = {Effect of Amyloid Beta on Cholesterol Metabolism-Correlated microRNAs in Primary Cultured Astrocytes of C57BL/6J Mice: A Focus on CYP46A1 and APOE Genes.},
journal = {Cell journal},
volume = {26},
number = {11},
pages = {625-631},
doi = {10.22074/cellj.2025.2029261.1587},
pmid = {40235143},
issn = {2228-5814},
abstract = {OBJECTIVE: The accumulation of amyloid plaques and disturbance of cholesterol homeostasis are implicated in the pathophysiology of Alzheimer's disease. Apolipoprotein E (ApoE) and cholesterol 24-hydroxylase (CYP46A1) are key proteins involved in the efflux and metabolism of excess cholesterol, and small non-coding RNAs (miRNAs), can help to regulate the expression of the genes encoding these proteins. The aim of the present study was to investigate the alterations in the expression of APOE and CYP46A1 genes, as well as their respective regulatory miRNAs, in astrocytes treated with amyloid beta (Aβ).
MATERIALS AND METHODS: In this experimental study, isolated astrocyte cells were cultured and treated with Aβ for 24 hours. Changes in the expression of APOE and CYP46A1 genes, as well as their regulating miRNAs, were assessed using the realtime polymerase chain reaction (PCR) technique.
RESULTS: The expression of APOE and CYP46A1 genes increased with Aβ treatment. MiR-33a-5p, as the negative regulator of the APOE gene exhibited significant decrease. Additionally, miR-let-7a-5p, as the positive regulator of the APOE gene, showed an increase in the Aβ treated group. Moreover, miR-98-5p, as the negative regulator of the CYP46A1 gene, showed a half-fold decrease. While, miR-27a-3p as the positive regulator of the CYP46A1 gene, increased significantly with Aβ treatment.
CONCLUSION: Alterations in the expression of APOE and CYP46A1 genes, as well as the expression of miRNAs regulating these genes, in astrocytes treated with Aβ suggests that the cell is attempting to modify the regulatory pathways of cholesterol homeostasis in the brain under pathological conditions, such as Alzheimer's disease.},
}
RevDate: 2025-04-16
CmpDate: 2025-04-16
The CentiMarker project: Standardizing quantitative Alzheimer's disease fluid biomarkers for biologic interpretation.
Alzheimer's & dementia : the journal of the Alzheimer's Association, 21(4):e14587.
INTRODUCTION: Biomarkers play a crucial role in understanding Alzheimer's disease (AD) pathogenesis and treatment effects. However, comparing biomarker measures without standardization and appreciating their magnitude relative to the disease can be challenging.
METHODS: To address this issue, we propose the CentiMarker approach, similar to Centiloid, which provides a standardized scale between normal (0) and nearly maximum abnormal AD (100) ranges. We applied this scale to cerebrospinal fluid (CSF) biomarkers in dominantly inherited AD and sporadic AD cohorts.
RESULTS: CentiMarkers facilitated the interpretation of disease abnormality, demonstrating comparable changes and distributions of AD biomarkers across disease stages. CentiMarkers make the treatment effect more comparable than their original scales across various biomarkers.
DISCUSSION: The versatility of CentiMarkers makes it a valuable tool for standardized biomarker comparison in AD research, enabling informed cross-study comparisons and contributing to accelerated therapeutic development. Adoption of the CentiMarker scale could enhance biomarker reporting and advance our understanding of AD.
HIGHLIGHTS: Comparing fluid biomarkers without appreciating their magnitude relative to the disease can be challenging. We propose a CentiMarker metric to standardize biomarker measures from normal (0) and nearly maximum abnormal AD (100) ranges. CentiMarkers make the treatment effect more comparable across various biomarkers than when using the original scales.
Additional Links: PMID-40235082
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40235082,
year = {2025},
author = {Wang, G and Li, Y and Xiong, C and Cao, Y and Schindler, SE and McDade, E and Blennow, K and Hansson, O and Dage, JL and Jack, CR and Teunissen, CE and Shaw, LM and Zetterberg, H and Ibanez, L and Timsina, J and Carlos, C and , and Bateman, RJ},
title = {The CentiMarker project: Standardizing quantitative Alzheimer's disease fluid biomarkers for biologic interpretation.},
journal = {Alzheimer's & dementia : the journal of the Alzheimer's Association},
volume = {21},
number = {4},
pages = {e14587},
doi = {10.1002/alz.14587},
pmid = {40235082},
issn = {1552-5279},
support = {/AG/NIA NIH HHS/United States ; U01 AG024904/NH/NIH HHS/United States ; U01AG042791-S1/NH/NIH HHS/United States ; R01AG046179//FNIH and Accelerating Medicines Partnership/ ; R01AG053267-S1//FNIH and Accelerating Medicines Partnership/ ; /ALZ/Alzheimer's Association/United States ; //Eli Lilly and Company/ ; //F. Hoffman-LaRoche Ltd./ ; //Avid Radiopharmaceuticals/ ; //GHR Foundation/ ; //The Dominantly Inherited Alzheimer Network/ ; //National Institute on Aging (NIA)/ ; //German Center for Neurodegenerative Diseases (DZNE)/ ; //Raul Carrea Institute for Neurological Research (FLENI)/ ; //Japan Agency for Medical Research and Development (AMED)/ ; //Korea Health Technology R&D/ ; //Korea Health Industry Development Institute (KHIDI)/ ; //Korea Dementia Research Center (KDRC)/ ; HI21C0066//the Ministry of Health & Welfare and Ministry of Science and ICT, Republic of Korea/ ; //Spanish Institute of Health Carlos III (ISCIII)/ ; //Alzheimer's Disease Neuroimaging Initiative/ ; W81XWH-12-2-0012//Department of Defense/ ; /EB/NIBIB NIH HHS/United States ; //AbbVie, Alzheimer's Association; Alzheimer's Drug Discovery Foundation; Araclon Biotech; BioClinica, Inc.; Biogen; Bristol-Myers Squibb Company; CereSpir, Inc.; Cogstate; Eisai Inc.; Elan Pharmaceuticals, Inc.; Eli Lilly and Company; EuroImmun; F. Hoffmann-La Roche Ltd/ ; //Genentech, Inc.; Fujirebio; GE Healthcare; IXICO Ltd.; Janssen Alzheimer Immunotherapy Research & Development, LLC.; Johnson & Johnson Pharmaceutical Research & Development LLC.; Lumosity; Lundbeck; Merck & Co., Inc.; Meso Scale Diagnostics, LLC.; NeuroRx Research; Neurotrack Technologies; Novartis Pharmaceuticals Corporation; Pfizer Inc.; Piramal Imaging; Servier; Takeda Pharmaceutical Company; and Transition Therapeutics. The Canadian Institutes of Health Research/ ; U01AG052564//Foundation for the National Institutes of Health/ ; },
mesh = {Humans ; *Alzheimer Disease/cerebrospinal fluid/diagnosis/genetics ; *Biomarkers/cerebrospinal fluid ; tau Proteins/cerebrospinal fluid ; Female ; Amyloid beta-Peptides/cerebrospinal fluid ; Male ; Aged ; Peptide Fragments/cerebrospinal fluid ; },
abstract = {INTRODUCTION: Biomarkers play a crucial role in understanding Alzheimer's disease (AD) pathogenesis and treatment effects. However, comparing biomarker measures without standardization and appreciating their magnitude relative to the disease can be challenging.
METHODS: To address this issue, we propose the CentiMarker approach, similar to Centiloid, which provides a standardized scale between normal (0) and nearly maximum abnormal AD (100) ranges. We applied this scale to cerebrospinal fluid (CSF) biomarkers in dominantly inherited AD and sporadic AD cohorts.
RESULTS: CentiMarkers facilitated the interpretation of disease abnormality, demonstrating comparable changes and distributions of AD biomarkers across disease stages. CentiMarkers make the treatment effect more comparable than their original scales across various biomarkers.
DISCUSSION: The versatility of CentiMarkers makes it a valuable tool for standardized biomarker comparison in AD research, enabling informed cross-study comparisons and contributing to accelerated therapeutic development. Adoption of the CentiMarker scale could enhance biomarker reporting and advance our understanding of AD.
HIGHLIGHTS: Comparing fluid biomarkers without appreciating their magnitude relative to the disease can be challenging. We propose a CentiMarker metric to standardize biomarker measures from normal (0) and nearly maximum abnormal AD (100) ranges. CentiMarkers make the treatment effect more comparable across various biomarkers than when using the original scales.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alzheimer Disease/cerebrospinal fluid/diagnosis/genetics
*Biomarkers/cerebrospinal fluid
tau Proteins/cerebrospinal fluid
Female
Amyloid beta-Peptides/cerebrospinal fluid
Male
Aged
Peptide Fragments/cerebrospinal fluid
RevDate: 2025-04-15
Design and synthesis of new 1,2,3-triazole-methoxyphenyl-1,3,4-oxadiazole derivatives: selective butyrylcholinesterase inhibitors against Alzheimer's disease.
BMC chemistry, 19(1):97.
Alzheimer's disease (AD) remains a significant public health challenge due to its progressive cognitive impairment and the absence of proven treatments. In this study, several novel 1,2,3-triazole-methoxyphenyl-1,3,4-oxadiazole derivatives were synthesized and evaluated for their ability to inhibit key enzymes associated with AD: acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). Structure-activity relationship (SAR) analysis revealed that derivatives featuring electron-withdrawing groups, particularly nitro and fluorine substituents, exhibited remarkable inhibitory activity against BChE while showing minimal effectiveness against AChE. Among these, compound 13s (R = 4-CH3, R' = 4-NO2) demonstrated the highest potency, selectively targeting BChE with an IC50 value of 11.01 µM. Molecular docking and molecular dynamics (MD) simulations provided deeper insights into the favorable interactions between these compounds and BChE. Additionally, cytotoxicity studies confirmed the active compound's limited toxicity toward normal cells, indicating a promising therapeutic profile. These findings suggest that the synthesized selective anti-BChE compounds hold potential for consideration in the later stages of AD treatment.
Additional Links: PMID-40234998
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40234998,
year = {2025},
author = {Iraji, A and Hariri, R and Hashempur, MH and Ghasemi, M and Pourtaher, H and Saeedi, M and Akbarzadeh, T},
title = {Design and synthesis of new 1,2,3-triazole-methoxyphenyl-1,3,4-oxadiazole derivatives: selective butyrylcholinesterase inhibitors against Alzheimer's disease.},
journal = {BMC chemistry},
volume = {19},
number = {1},
pages = {97},
pmid = {40234998},
issn = {2661-801X},
abstract = {Alzheimer's disease (AD) remains a significant public health challenge due to its progressive cognitive impairment and the absence of proven treatments. In this study, several novel 1,2,3-triazole-methoxyphenyl-1,3,4-oxadiazole derivatives were synthesized and evaluated for their ability to inhibit key enzymes associated with AD: acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). Structure-activity relationship (SAR) analysis revealed that derivatives featuring electron-withdrawing groups, particularly nitro and fluorine substituents, exhibited remarkable inhibitory activity against BChE while showing minimal effectiveness against AChE. Among these, compound 13s (R = 4-CH3, R' = 4-NO2) demonstrated the highest potency, selectively targeting BChE with an IC50 value of 11.01 µM. Molecular docking and molecular dynamics (MD) simulations provided deeper insights into the favorable interactions between these compounds and BChE. Additionally, cytotoxicity studies confirmed the active compound's limited toxicity toward normal cells, indicating a promising therapeutic profile. These findings suggest that the synthesized selective anti-BChE compounds hold potential for consideration in the later stages of AD treatment.},
}
RevDate: 2025-04-15
Huang-Lian-Jie-Du decoction alleviates cognitive deficits in Alzheimer's disease model 5xFAD mice by inhibiting Trem2/Dap12 signaling pathway.
Chinese medicine, 20(1):50.
BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder predominantly affecting the elderly population. It is characterized by cognitive deficits associated with the accumulation of amyloid-beta plaques and neurofibrillary tangles. Huang-Lian-Jie-Du (HLJD) decoction, recognized as a representative formulation with heat-clearing and detoxification effects, has been demonstrated to be effective in treating AD. However, the underlying mechanisms require further investigation.
METHODS: 5xFAD mice were administrated low and high doses of HLJD. The Morris water maze test was conducted to assess the effects of HLJD. Aβ42 and total tau protein levels were evaluated. Additionally, network pharmacology analysis was performed to identify therapeutic targets of HLJD's active components and their relevance to AD. ELISA, qPCR, Western Blot, and immunofluorescence assays were employed to confirm the identified pathways. Finally, primary microglia isolated from 5xFAD mice were used to validate the candidate targets of HLJD.
RESULTS: HLJD improved cognitive deficits in 5xFAD mice and reduced amyloid plaque deposition and tau protein levels. Network pharmacology analysis indicated that HLJD influences the neuroinflammatory response, particularly through the Dap12 signaling pathway. This was confirmed by reduced levels of neuroinflammation markers, including TNF-α, IL-1β, IL-6, and indicators of microglial activation and polarization. The expression of Trem2 and Dap12 in the hippocampus (HIP) of 5xFAD mice, as well as in the isolated primary microglia, were downregulated following HLJD treatment.
CONCLUSION: Our study indicates that HLJD alleviates cognitive deficits in AD by suppressing the Trem2/Dap12 signaling pathway in the HIP of 5xFAD mice, thereby inhibiting microglial neuroinflammation.
Additional Links: PMID-40234956
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40234956,
year = {2025},
author = {Pang, RK and Shi, J and Peng, XY and Su, S and Zheng, JY and Le, K and Keng, VW and Zhang, SJ and Li, XX},
title = {Huang-Lian-Jie-Du decoction alleviates cognitive deficits in Alzheimer's disease model 5xFAD mice by inhibiting Trem2/Dap12 signaling pathway.},
journal = {Chinese medicine},
volume = {20},
number = {1},
pages = {50},
pmid = {40234956},
issn = {1749-8546},
abstract = {BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder predominantly affecting the elderly population. It is characterized by cognitive deficits associated with the accumulation of amyloid-beta plaques and neurofibrillary tangles. Huang-Lian-Jie-Du (HLJD) decoction, recognized as a representative formulation with heat-clearing and detoxification effects, has been demonstrated to be effective in treating AD. However, the underlying mechanisms require further investigation.
METHODS: 5xFAD mice were administrated low and high doses of HLJD. The Morris water maze test was conducted to assess the effects of HLJD. Aβ42 and total tau protein levels were evaluated. Additionally, network pharmacology analysis was performed to identify therapeutic targets of HLJD's active components and their relevance to AD. ELISA, qPCR, Western Blot, and immunofluorescence assays were employed to confirm the identified pathways. Finally, primary microglia isolated from 5xFAD mice were used to validate the candidate targets of HLJD.
RESULTS: HLJD improved cognitive deficits in 5xFAD mice and reduced amyloid plaque deposition and tau protein levels. Network pharmacology analysis indicated that HLJD influences the neuroinflammatory response, particularly through the Dap12 signaling pathway. This was confirmed by reduced levels of neuroinflammation markers, including TNF-α, IL-1β, IL-6, and indicators of microglial activation and polarization. The expression of Trem2 and Dap12 in the hippocampus (HIP) of 5xFAD mice, as well as in the isolated primary microglia, were downregulated following HLJD treatment.
CONCLUSION: Our study indicates that HLJD alleviates cognitive deficits in AD by suppressing the Trem2/Dap12 signaling pathway in the HIP of 5xFAD mice, thereby inhibiting microglial neuroinflammation.},
}
RevDate: 2025-04-15
Functionalized Nanoparticles: A Promising Approach for Effective Management of Alzheimer's Disease.
Molecular neurobiology [Epub ahead of print].
The severe neurodegenerative disease known as Alzheimer's disease (AD) is typified by a progressive loss of memory and cognitive function. The prevalence of AD is rising due to an aging global population, calling for novel treatment strategies. A potential treatment option for AD that shows promise is the use of functionalized nanoparticles (NPs). Recent developments in the synthesis, design, and use of functionalized NPs in AD therapy are examined in this review. An outline of the pathophysiological mechanisms underlying AD is given in the first section, focusing on the roles played by tau protein aggregates and amyloid-beta plaques in the development of the illness. We then explore the many approaches used to functionalize NPs, such as surface alterations and bioconjugation methods, which enable accurate drug administration, targeted delivery, and enhanced biocompatibility. The review also emphasizes the therapeutic potential of functionalized NPs, highlighting their capacity to improve neuroprotection, lower amyloid-beta aggregation, and improve blood-brain barrier penetration. The potential of NPs as a tool for disease modification and symptom relief is highlighted by recent pre-clinical and clinical research. Concerns about toxicity and safety are also covered, underscoring the significance of thorough testing and the field's future directions. Functionalized NPs have great promise as a multimodal strategy to treat AD, offering patients hope for better quality of life, early diagnosis, and efficient disease treatment. This study highlights the growing role of nanotechnology in the search for novel and potent therapies for AD.
Additional Links: PMID-40234291
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40234291,
year = {2025},
author = {Bogadi, S and Bhaskaran, M and Ravichandran, V and Nesamony, J and Chelliah, S and Kuppusamy, G and Prakash, GM and Karri, VVSR and Mallick, S and Farahim, F and Ali, T and Babu, DR and Subramaniyan, V},
title = {Functionalized Nanoparticles: A Promising Approach for Effective Management of Alzheimer's Disease.},
journal = {Molecular neurobiology},
volume = {},
number = {},
pages = {},
pmid = {40234291},
issn = {1559-1182},
abstract = {The severe neurodegenerative disease known as Alzheimer's disease (AD) is typified by a progressive loss of memory and cognitive function. The prevalence of AD is rising due to an aging global population, calling for novel treatment strategies. A potential treatment option for AD that shows promise is the use of functionalized nanoparticles (NPs). Recent developments in the synthesis, design, and use of functionalized NPs in AD therapy are examined in this review. An outline of the pathophysiological mechanisms underlying AD is given in the first section, focusing on the roles played by tau protein aggregates and amyloid-beta plaques in the development of the illness. We then explore the many approaches used to functionalize NPs, such as surface alterations and bioconjugation methods, which enable accurate drug administration, targeted delivery, and enhanced biocompatibility. The review also emphasizes the therapeutic potential of functionalized NPs, highlighting their capacity to improve neuroprotection, lower amyloid-beta aggregation, and improve blood-brain barrier penetration. The potential of NPs as a tool for disease modification and symptom relief is highlighted by recent pre-clinical and clinical research. Concerns about toxicity and safety are also covered, underscoring the significance of thorough testing and the field's future directions. Functionalized NPs have great promise as a multimodal strategy to treat AD, offering patients hope for better quality of life, early diagnosis, and efficient disease treatment. This study highlights the growing role of nanotechnology in the search for novel and potent therapies for AD.},
}
RevDate: 2025-04-15
Magnesium-L-threonate Ameliorates Cognitive Deficit by Attenuating Adult Hippocampal Neurogenesis Impairment in a Mouse Model of Alzheimer's Disease.
Experimental neurobiology pii:en24030 [Epub ahead of print].
Impaired adult hippocampal neurogenesis is a key pathological mechanism contributing to memory deficits in Alzheimer's disease (AD). Recent studies have shown that elevating magnesium levels promotes neurogenesis by enhancing the neuronal differentiation of adult neural progenitor cells in vitro. Therefore, this in vivo study aims to determine if magnesium-L-threonate (MgT) can ameliorate cognitive deficit of AD mice by attenuating adult hippocampal neurogenesis impairment and to reveal the underlying mechanisms. APPswe/PS1dE9 mice were treated with different doses of MgT and ERK inhibitor PD0325901. The memory ability of each mouse was recorded by Morris Water Maze test. After cognitive test, hippocampus tissues were collected to measure the proportion of BrdU/doublecortin double-labeled cells using the flow cytometry test and assess the expression of doublecortin using PCR and Western blot. Furthermore, the activations of CREB, ERK, P38 and JNK were measured by Western blot to identify the involved mechanisms. The cognitive test confirmed that MgT treatment attenuated the memory impairment of APPswe/PS1dE9 mice. Flow cytometry test showed that Brdu/doublecortin labeled newborn neurons gradually increased following MgT administration. In line with the flow cytometry results, Western blot and PCR confirmed that MgT administration significantly increased doublecortin expression levels. Furthermore, the ratios of p-ERK/ERK and p-CREB/CREB increased with MgT elevation. In addition, these effects of MgT treatment were markedly reversed by PD0325901 supplementation. In conclusion, MgT treatment improved cognitive decline by ameliorating adult hippocampal neurogenesis impairment in this AD model, possibly via ERK/CREB activation.
Additional Links: PMID-40234095
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40234095,
year = {2025},
author = {Xiong, Y and Yang, Y and Ruan, Y and Ou, W and Hu, Z and Li, W and Xiao, N and Liao, W and Liu, J and Liu, Z and Luo, Q and Liu, F and Liu, J},
title = {Magnesium-L-threonate Ameliorates Cognitive Deficit by Attenuating Adult Hippocampal Neurogenesis Impairment in a Mouse Model of Alzheimer's Disease.},
journal = {Experimental neurobiology},
volume = {},
number = {},
pages = {},
doi = {10.5607/en24030},
pmid = {40234095},
issn = {1226-2560},
abstract = {Impaired adult hippocampal neurogenesis is a key pathological mechanism contributing to memory deficits in Alzheimer's disease (AD). Recent studies have shown that elevating magnesium levels promotes neurogenesis by enhancing the neuronal differentiation of adult neural progenitor cells in vitro. Therefore, this in vivo study aims to determine if magnesium-L-threonate (MgT) can ameliorate cognitive deficit of AD mice by attenuating adult hippocampal neurogenesis impairment and to reveal the underlying mechanisms. APPswe/PS1dE9 mice were treated with different doses of MgT and ERK inhibitor PD0325901. The memory ability of each mouse was recorded by Morris Water Maze test. After cognitive test, hippocampus tissues were collected to measure the proportion of BrdU/doublecortin double-labeled cells using the flow cytometry test and assess the expression of doublecortin using PCR and Western blot. Furthermore, the activations of CREB, ERK, P38 and JNK were measured by Western blot to identify the involved mechanisms. The cognitive test confirmed that MgT treatment attenuated the memory impairment of APPswe/PS1dE9 mice. Flow cytometry test showed that Brdu/doublecortin labeled newborn neurons gradually increased following MgT administration. In line with the flow cytometry results, Western blot and PCR confirmed that MgT administration significantly increased doublecortin expression levels. Furthermore, the ratios of p-ERK/ERK and p-CREB/CREB increased with MgT elevation. In addition, these effects of MgT treatment were markedly reversed by PD0325901 supplementation. In conclusion, MgT treatment improved cognitive decline by ameliorating adult hippocampal neurogenesis impairment in this AD model, possibly via ERK/CREB activation.},
}
RevDate: 2025-04-15
CmpDate: 2025-04-15
Vigna radiata extracts in pumpkin and soya bean oil: A novel therapeutic approach for Alzheimer's disease.
PloS one, 20(4):e0321183 pii:PONE-D-24-45075.
Vigna radiate also known as mung beans, contains various bioactive compounds like polyphenols, flavonoids, and saponins. V. radiata therapeutic potential is enhanced by preparation of its extract in Pumpkin oil and soya bean oil by enrichment of bioactive compounds holding antioxidant, anti-inflammatory, and neuro-protective properties. The research study was aimed was to explore the healing endeavors of V. radiate pumpkin and soya bean oil extract in rectification of neuro-motor dysfunction and mental health decline in Alzheimer's disease (AD) rat model. After preliminary physico-phytochemical characterization and GC-MS analysis, AD model was established by administration of oral D-galactose and aluminum chloride 150 mg/kg each for 42 days daily. V. radiate extract in pumpkin and soya bean oil at doses 250 and 500 mg/kg was administered and rivastigmine (3 milligrams per kilogram) to treatment animals. To determine the cognitive decline and neuro-coordination dysfunctions behavioral tests were performed along with biochemical, neurochemical and histopathological analysis. ELISA and real time polymerase chain reaction were carried out to estimate the expression of tumor necrosis factor-α, Interleukine-6 and mRNA expression of neurodegenerative biomarkers. Gas chromatography Mass Spectrometry findings revealed the existence of favorable amount of neuro-defensive bioactive compounds in both oil extracts.V. radiate pumpkin and soya bean oil extract dose proportionally alleviated the behavioral dysfunctions, modulated the first line antioxidant enzymes and neurotransmitters s' level with anticholinesterase pursuits. The mRNA expression of AChE, IL-1β, TNF-α, IL-1α and β secretase were downregulated by these extracts treatment. V. radiate oil extracts also modulated the neuro-inflammatory protein expression and histopathological hallmarks in AD model animals. Therefore, it is purposed that V. radiate enriched extract in pumpkin and soya bean oil could be used to treat AD like memory dysfunction and motor symptoms.
Additional Links: PMID-40233108
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40233108,
year = {2025},
author = {Amin, H and Bukhari, SA and Chauhdary, Z and Akhter, N and Saleem, M},
title = {Vigna radiata extracts in pumpkin and soya bean oil: A novel therapeutic approach for Alzheimer's disease.},
journal = {PloS one},
volume = {20},
number = {4},
pages = {e0321183},
doi = {10.1371/journal.pone.0321183},
pmid = {40233108},
issn = {1932-6203},
mesh = {Animals ; *Alzheimer Disease/drug therapy/metabolism/pathology/chemically induced ; *Cucurbita/chemistry ; Rats ; *Plant Extracts/pharmacology/therapeutic use/chemistry ; *Soybean Oil/chemistry ; *Vigna/chemistry ; Male ; Disease Models, Animal ; Rats, Wistar ; Antioxidants ; Neuroprotective Agents/pharmacology ; },
abstract = {Vigna radiate also known as mung beans, contains various bioactive compounds like polyphenols, flavonoids, and saponins. V. radiata therapeutic potential is enhanced by preparation of its extract in Pumpkin oil and soya bean oil by enrichment of bioactive compounds holding antioxidant, anti-inflammatory, and neuro-protective properties. The research study was aimed was to explore the healing endeavors of V. radiate pumpkin and soya bean oil extract in rectification of neuro-motor dysfunction and mental health decline in Alzheimer's disease (AD) rat model. After preliminary physico-phytochemical characterization and GC-MS analysis, AD model was established by administration of oral D-galactose and aluminum chloride 150 mg/kg each for 42 days daily. V. radiate extract in pumpkin and soya bean oil at doses 250 and 500 mg/kg was administered and rivastigmine (3 milligrams per kilogram) to treatment animals. To determine the cognitive decline and neuro-coordination dysfunctions behavioral tests were performed along with biochemical, neurochemical and histopathological analysis. ELISA and real time polymerase chain reaction were carried out to estimate the expression of tumor necrosis factor-α, Interleukine-6 and mRNA expression of neurodegenerative biomarkers. Gas chromatography Mass Spectrometry findings revealed the existence of favorable amount of neuro-defensive bioactive compounds in both oil extracts.V. radiate pumpkin and soya bean oil extract dose proportionally alleviated the behavioral dysfunctions, modulated the first line antioxidant enzymes and neurotransmitters s' level with anticholinesterase pursuits. The mRNA expression of AChE, IL-1β, TNF-α, IL-1α and β secretase were downregulated by these extracts treatment. V. radiate oil extracts also modulated the neuro-inflammatory protein expression and histopathological hallmarks in AD model animals. Therefore, it is purposed that V. radiate enriched extract in pumpkin and soya bean oil could be used to treat AD like memory dysfunction and motor symptoms.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Alzheimer Disease/drug therapy/metabolism/pathology/chemically induced
*Cucurbita/chemistry
Rats
*Plant Extracts/pharmacology/therapeutic use/chemistry
*Soybean Oil/chemistry
*Vigna/chemistry
Male
Disease Models, Animal
Rats, Wistar
Antioxidants
Neuroprotective Agents/pharmacology
RevDate: 2025-04-15
Harnessing the Potential of Walnut Leaves from Nerpio: Unveiling Extraction Techniques and Bioactivity Through Caenorhabditis elegans Studies.
Foods (Basel, Switzerland), 14(6): pii:foods14061048.
This study used Juglans regia leaves from the Gran Jefe variety; this indigenous cultivar from Nerpio is highly valued for its quality and distinct characteristics. This type of walnut is traditionally cultivated in the region and is noted for its organoleptic properties and adaptation to local climatic conditions. Two solvents were tested to determine the optimal extraction conditions for phenolic compounds: 80% ethanol and water. Direct homogenization with an Ultra-Turrax, direct ultrasound, and indirect ultrasound treatments were compared for ethanol extraction. Water extractions were conducted using direct and indirect ultrasound, infusion, and decoction. Compared to water extraction, 80% ethanol proved to be more efficient. Extracting phenolic compounds from 'Gran Jefe' walnut leaves was most effective when using direct extraction methods without either ultrasound assistance or indirect ultrasound treatment. The main compounds identified were trans-3-caffeoylquinic acid and quercetin-3-hexoside isomer 1. The ethanolic extract obtained through direct extraction was selected to study further the bioactivities of 'Gran Jefe' walnut leaves using C. elegans as an in vivo model. Results indicated that the leaf extract enhanced thermal and oxidative stress resistance, promoted fertility, and exhibited neuroprotective effects in models of Alzheimer's and Parkinson's diseases. The observed bioactivities were attributed to the free phenolics present in the ethanolic extract.
Additional Links: PMID-40232090
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40232090,
year = {2025},
author = {Hamdi, A and Córdoba-Rojano, MA and Monje-Moreno, JM and Guillén-Izquierdo, E and Rodríguez-Arcos, R and Jiménez-Araujo, A and Muñoz-Ruiz, MJ and Guillén-Bejarano, R},
title = {Harnessing the Potential of Walnut Leaves from Nerpio: Unveiling Extraction Techniques and Bioactivity Through Caenorhabditis elegans Studies.},
journal = {Foods (Basel, Switzerland)},
volume = {14},
number = {6},
pages = {},
doi = {10.3390/foods14061048},
pmid = {40232090},
issn = {2304-8158},
support = {research contract number 20232315//Asociación de Productores de Nuez de Nerpio/ ; },
abstract = {This study used Juglans regia leaves from the Gran Jefe variety; this indigenous cultivar from Nerpio is highly valued for its quality and distinct characteristics. This type of walnut is traditionally cultivated in the region and is noted for its organoleptic properties and adaptation to local climatic conditions. Two solvents were tested to determine the optimal extraction conditions for phenolic compounds: 80% ethanol and water. Direct homogenization with an Ultra-Turrax, direct ultrasound, and indirect ultrasound treatments were compared for ethanol extraction. Water extractions were conducted using direct and indirect ultrasound, infusion, and decoction. Compared to water extraction, 80% ethanol proved to be more efficient. Extracting phenolic compounds from 'Gran Jefe' walnut leaves was most effective when using direct extraction methods without either ultrasound assistance or indirect ultrasound treatment. The main compounds identified were trans-3-caffeoylquinic acid and quercetin-3-hexoside isomer 1. The ethanolic extract obtained through direct extraction was selected to study further the bioactivities of 'Gran Jefe' walnut leaves using C. elegans as an in vivo model. Results indicated that the leaf extract enhanced thermal and oxidative stress resistance, promoted fertility, and exhibited neuroprotective effects in models of Alzheimer's and Parkinson's diseases. The observed bioactivities were attributed to the free phenolics present in the ethanolic extract.},
}
RevDate: 2025-04-15
Advance Care Planning and Unlimited Treatment Preferences in Dementia Scenarios: Insights From Community-Dwelling Adults.
The American journal of hospice & palliative care [Epub ahead of print].
Introduction: Dementia leads to progressive cognitive decline, impairing self-care and decision making. Advance directives (AdvDirs) enable individuals to document healthcare preferences while cognitively capable, ensuring value-aligned care and reducing caregiver burden. This study explores factors influencing preferences for unlimited medical treatment in hypothetical Alzheimer's disease/dementia scenarios among community-dwelling adults. Methods: This cross-sectional study surveyed 163 community-dwelling adults (18+), using structured questionnaires to collect sociodemographic, health, and AdvDir-related data. Key predictors included attitudes toward life-sustaining treatments, comfort discussing death, religious practices, and interest in quality-of-life information related to end-of-life care. The primary outcome was preference for unlimited medical treatment in hypothetical dementia scenarios. Bivariate and multivariate logistic analyses assessed associations, adjusting for covariates. Results: In the dementia scenario, 26.9% of participants preferred unlimited medical treatment. This preference was strongly associated with a pre-existing attitude favoring life-sustaining treatments (OR = 4.24, 95% CI: 1.73 - 10.37, P = 0.002) and religious beliefs (OR = 5.68, 95% CI: 1.51-21.43, P = 0.01). Conversely, an interest in learning about quality of life at the end of life was negatively associated with preferring unlimited treatment (OR = 0.29, 95% CI: 0.09-0.89, P = 0.03). Discussion: Our findings highlight the need to align advance care planning with individuals' values, beliefs, and religious practices. Raising awareness of quality-of-life considerations in end-of-life care may lead to a shift in preference toward palliative care rather than aggressive treatment. Healthcare providers should discuss treatment trade-offs with cultural and religious sensitivity to support informed decision making.
Additional Links: PMID-40231700
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40231700,
year = {2025},
author = {Young, Y and Liu, Y and Tu, Y and Chiu, WY and Shayya, A and O'Grady, T},
title = {Advance Care Planning and Unlimited Treatment Preferences in Dementia Scenarios: Insights From Community-Dwelling Adults.},
journal = {The American journal of hospice & palliative care},
volume = {},
number = {},
pages = {10499091251334090},
doi = {10.1177/10499091251334090},
pmid = {40231700},
issn = {1938-2715},
abstract = {Introduction: Dementia leads to progressive cognitive decline, impairing self-care and decision making. Advance directives (AdvDirs) enable individuals to document healthcare preferences while cognitively capable, ensuring value-aligned care and reducing caregiver burden. This study explores factors influencing preferences for unlimited medical treatment in hypothetical Alzheimer's disease/dementia scenarios among community-dwelling adults. Methods: This cross-sectional study surveyed 163 community-dwelling adults (18+), using structured questionnaires to collect sociodemographic, health, and AdvDir-related data. Key predictors included attitudes toward life-sustaining treatments, comfort discussing death, religious practices, and interest in quality-of-life information related to end-of-life care. The primary outcome was preference for unlimited medical treatment in hypothetical dementia scenarios. Bivariate and multivariate logistic analyses assessed associations, adjusting for covariates. Results: In the dementia scenario, 26.9% of participants preferred unlimited medical treatment. This preference was strongly associated with a pre-existing attitude favoring life-sustaining treatments (OR = 4.24, 95% CI: 1.73 - 10.37, P = 0.002) and religious beliefs (OR = 5.68, 95% CI: 1.51-21.43, P = 0.01). Conversely, an interest in learning about quality of life at the end of life was negatively associated with preferring unlimited treatment (OR = 0.29, 95% CI: 0.09-0.89, P = 0.03). Discussion: Our findings highlight the need to align advance care planning with individuals' values, beliefs, and religious practices. Raising awareness of quality-of-life considerations in end-of-life care may lead to a shift in preference toward palliative care rather than aggressive treatment. Healthcare providers should discuss treatment trade-offs with cultural and religious sensitivity to support informed decision making.},
}
▼ ▼ 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.