Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (221)

Search Parameters:
Keywords = neuropathological alterations

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 3482 KB  
Article
Neuroprotective Effects and Mechanisms of Arecoline Against H2O2-Induced Damage in SH-SY5Y Cells
by Xiangfei Zhang, Jingwen Cui, Jing Sun, Fengzhong Wang, Bei Fan and Cong Lu
Int. J. Mol. Sci. 2025, 26(21), 10355; https://doi.org/10.3390/ijms262110355 - 24 Oct 2025
Viewed by 277
Abstract
An overproduction of reactive oxygen species (ROS) creates oxidative stress that disrupts neuronal activity and contributes to the pathogenesis of neurodegenerative diseases. Arecoline, the predominant alkaloid component of Areca catechu L., is known for multiple biological activities, yet its involvement in neuronal oxidative [...] Read more.
An overproduction of reactive oxygen species (ROS) creates oxidative stress that disrupts neuronal activity and contributes to the pathogenesis of neurodegenerative diseases. Arecoline, the predominant alkaloid component of Areca catechu L., is known for multiple biological activities, yet its involvement in neuronal oxidative injury has not been fully clarified. This study investigated arecoline’s effect on hydrogen peroxide (H2O2)-induced toxicity in SH-SY5Y human neuroblastoma cells (SH-SY5Y). Arecoline pretreatment significantly improved cell viability and preserved plasma membrane integrity, accompanied by reduced lipid peroxidation and restoration of cellular antioxidant enzyme activities. Moreover, arecoline maintained mitochondrial membrane potential and suppressed apoptotic progression. At the molecular level, Arecoline stimulated nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) protein expression, concurrently diminishing Kelch-like ECH-associated protein 1 (Keap1) levels. In parallel, it altered the apoptosis profile by increasing B-cell lymphoma 2 (Bcl2) levels and decreasing Bcl-2-associated X protein (Bax) and total cysteine aspartate protease-3 (Caspase-3) protein expression. Collectively, the findings suggest that arecoline safeguards neurons against oxidative stress by simultaneously activating antioxidant defenses and restraining apoptosis. This study adds novel molecular evidence supporting the potential neuroprotective relevance of arecoline in oxidative stress-related neuropathology. Full article
(This article belongs to the Collection Advances in Cell and Molecular Biology)
Show Figures

Figure 1

17 pages, 877 KB  
Review
Synaptic Pathology in Traumatic Brain Injury and Therapeutic Insights
by Poojith Nuthalapati, Sophie E. Holmes, Hamada H. Altalib and Arman Fesharaki-Zadeh
Int. J. Mol. Sci. 2025, 26(19), 9604; https://doi.org/10.3390/ijms26199604 - 1 Oct 2025
Viewed by 853
Abstract
Traumatic brain injury (TBI) results in a cascade of neuropathological events, which can significantly disrupt synaptic integrity. This review explores the acute, subacute and chronic phases of synaptic dysfunction and loss in trauma which commence post-TBI, and their contribution to the subsequent neurological [...] Read more.
Traumatic brain injury (TBI) results in a cascade of neuropathological events, which can significantly disrupt synaptic integrity. This review explores the acute, subacute and chronic phases of synaptic dysfunction and loss in trauma which commence post-TBI, and their contribution to the subsequent neurological sequelae. Central to these disruptions is the loss of dendritic spines and impaired synaptic plasticity, which compromise neuronal connectivity and signal transmission. During the acute phase of TBI, mechanical injury triggers presynaptic glutamate secretion and Ca2+ ion-mediated excitotoxic injury, accompanied by cerebral edema, mitochondrial dysfunction and the loss of the mushroom-shaped architecture of the dendritic spines. The subacute phase is marked by continued glutamate excitotoxicity and GABAergic disruption, along with neuroinflammatory pathology and autophagy. In the chronic phase, long-term structural remodeling and reduced synaptic densities are evident. These chronic alterations underlie persistent cognitive and memory deficits, mood disturbances and the development of post-traumatic epilepsy. Understanding the phase-specific progression of TBI-related synaptic dysfunction is essential for targeted interventions. Novel therapeutic strategies primarily focus on how to effectively counter acute excitotoxicity and neuroinflammatory cascades. Future approaches may benefit from boosting synaptic repair and modulating neurotransmitter systems in a phase-specific manner, thereby mitigating the long-term impact of TBI on neuronal function. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

30 pages, 724 KB  
Review
Astrocytes and Astrocyte-Derived Extracellular Conduits in Opiate-Mediated Neurological Disorders
by Sudipta Ray, Souvik Datta, Arnab Saha and Susmita Sil
Cells 2025, 14(18), 1454; https://doi.org/10.3390/cells14181454 - 17 Sep 2025
Viewed by 1577
Abstract
Opioid-use disorder (OUD) poses a growing global health crisis, with chronic opioid exposure linked not only to addiction but also to enduring neurological impairments. While traditional research has focused primarily on neuronal alterations, emerging evidence underscores the pivotal role of astrocytes, abundant glial [...] Read more.
Opioid-use disorder (OUD) poses a growing global health crisis, with chronic opioid exposure linked not only to addiction but also to enduring neurological impairments. While traditional research has focused primarily on neuronal alterations, emerging evidence underscores the pivotal role of astrocytes, abundant glial cells in the central nervous system, and their secreted extracellular vesicles (EVs) in opioid-mediated neuropathology. This review delineates the mechanistic roles of astrocytes and astrocyte-derived EVs (ADEVs) across a spectrum of opioids, including morphine, heroin, fentanyl, codeine, tramadol, buprenorphine, and methadone. Opioids disrupt astrocytic homeostasis by impairing glutamate regulation, altering the redox balance, and activating pro-inflammatory signaling pathways. In response, astrocytes release EVs enriched with neurotoxic cargo, including amyloidogenic proteins, cytokines, microRNAs, and long non-coding RNAs, that propagate neuroinflammation, compromise blood–brain barrier (BBB) integrity, and exacerbate synaptic dysfunction. Preclinical models and in vitro studies reveal drug-specific astrocytic responses and ADEV profiles, implicating these vesicles in modulating microglial function, neuroimmune signaling, and neuronal viability. Notably, morphine-induced ADEVs promote amyloidosis and inflammatory signaling, while heroin and fentanyl affect glutamatergic and inflammasome pathways. Even opioids used in therapy, such as buprenorphine and methadone, alter astrocyte morphology and EV cargo, particularly during neurodevelopment. Collectively, these findings advance a neuro-glial paradigm for understanding opioid-induced brain injury and highlight ADEVs as both biomarkers and mediators of neuropathology. Targeting astrocyte-EV signaling pathways represents a promising therapeutic avenue to mitigate long-term neurological consequences of opioid exposure and improve outcomes in OUD. Full article
(This article belongs to the Special Issue The Role Glial Cells in Neurodegenerative Disorders)
Show Figures

Figure 1

22 pages, 3886 KB  
Article
Retrospective Analysis of Central Nervous System Diseases in Dogs, with Special Focus on Non-Suppurative Encephalomyelitis (1962–2022)
by Inga Marie Nägler, Adnan Fayyad, Christina Puff, Wolfgang Baumgärtner and Peter Wohlsein
Vet. Sci. 2025, 12(9), 869; https://doi.org/10.3390/vetsci12090869 - 8 Sep 2025
Viewed by 1369
Abstract
Studies spanning decades provide important information about the epidemiology and occurrence of a broad range of diseases that affect the central nervous system (CNS) of dogs. This study analyzed records and formalin-fixed paraffin-embedded (FFPE) CNS tissue samples from necropsied dogs with neuropathologic changes [...] Read more.
Studies spanning decades provide important information about the epidemiology and occurrence of a broad range of diseases that affect the central nervous system (CNS) of dogs. This study analyzed records and formalin-fixed paraffin-embedded (FFPE) CNS tissue samples from necropsied dogs with neuropathologic changes between 1962 and 2022. A total of 134,854 animals, including 20,117 dogs, were submitted for necropsy during this time span. Of these dogs, 2646 displayed alterations of the CNS. Degenerative and non-suppurative inflammatory lesions were the most common changes, accounting for 35.6% and 28.6%, respectively. Vascular diseases, neoplasms, congenital malformations, and suppurative inflammation represented 13.8%, 8.6%, 7.2%, and 5.4% of cases, respectively. Morbillivirus canis, the agent of canine distemper, was the most commonly diagnosed. The second most commonly detected virus, varicellovirus suidalpha1, the agent of pseudorabies, occurred almost exclusively between the mid-1970s and 1990s. Other pathogens, including Lyssavirus rabies, canine herpes virus, tick-borne encephalitis virus, apicomplexan parasites, such as Neospora caninum and Toxoplasma gondii, as well as fungal and other parasitic infections, were less frequently diagnosed. Interestingly, 47.6% of cases with non-suppurative inflammation remained etiologically undetermined. This study provides insights into the epidemiology of canine neurotropic infections and shows the value of FFPE material for investigations of past disease outbreaks. Full article
Show Figures

Figure 1

15 pages, 8949 KB  
Article
Protein Expression of TXNIP in the Dopaminergic Neurons of Subjects with Parkinson’s Disease: Evidence from a Pilot Study
by Francesca A. Schillaci, Giuseppe Lanza, Maria Grazia Salluzzo, Raffaele Ferri and Michele Salemi
Life 2025, 15(8), 1252; https://doi.org/10.3390/life15081252 - 7 Aug 2025
Viewed by 668
Abstract
Parkinson’s disease (PD) is a progressive, multisystemic α-synucleinopathy, recognized as the second most prevalent neurodegenerative disorder globally. Its neuropathology is characterized by the degeneration of dopaminergic neurons, particularly in the substantia nigra pars compacta (SNpc), and the intraneuronal accumulation of α-synuclein-forming Lewy bodies. [...] Read more.
Parkinson’s disease (PD) is a progressive, multisystemic α-synucleinopathy, recognized as the second most prevalent neurodegenerative disorder globally. Its neuropathology is characterized by the degeneration of dopaminergic neurons, particularly in the substantia nigra pars compacta (SNpc), and the intraneuronal accumulation of α-synuclein-forming Lewy bodies. Oxidative stress is a key contributor to PD pathogenesis. Thioredoxin-interacting protein (TXNIP) is a crucial regulator of cellular redox balance, inhibiting the antioxidant function of thioredoxin. This pilot study aimed to investigate the protein expression and localization of TXNIP in the SNpc of PD patients compared to healthy controls. We performed immunohistochemical analyses on 12 post-mortem human brain sections (formalin-fixed, paraffin-embedded) from six subjects with PD and six healthy controls. The study was performed on PD subjects with Braak stage 6. Our findings revealed that in control samples, TXNIP protein was distinctly and closely associated with neuromelanin (NM) pigment within the cytoplasm of SNpc dopaminergic neurons. Conversely, in PD samples, there was a markedly weak cytoplasmic expression of TXNIP, and critically, this association with NM pigment was absent. Furthermore, PD samples exhibited a significant reduction in both dopaminergic neurons and NM content, consistent with advanced disease. These findings, which mirror previous transcriptomic data showing TXNIP gene under-expression in the same subjects, suggest that altered TXNIP expression and localization in SNpc dopaminergic neurons are features of late-stage PD, potentially reflecting neuronal dysfunction and loss. Full article
(This article belongs to the Special Issue Regulation of Cellular Signaling Pathways in the Metabolic Syndrome)
Show Figures

Figure 1

22 pages, 9552 KB  
Article
Benefits of Maternal Choline Supplementation on Aged Basal Forebrain Cholinergic Neurons (BFCNs) in a Mouse Model of Down Syndrome and Alzheimer’s Disease
by Melissa J. Alldred, Harshitha Pidikiti, Kyrillos W. Ibrahim, Sang Han Lee, Adriana Heguy, Gabriela Chiosis, Elliott J. Mufson, Grace E. Stutzmann and Stephen D. Ginsberg
Biomolecules 2025, 15(8), 1131; https://doi.org/10.3390/biom15081131 - 5 Aug 2025
Viewed by 1221
Abstract
Down syndrome (DS), stemming from the triplication of human chromosome 21, results in intellectual disability, with early mid-life onset of Alzheimer’s disease (AD) pathology. Early interventions to reduce cognitive impairments and neuropathology are lacking. One modality, maternal choline supplementation (MCS), has shown beneficial [...] Read more.
Down syndrome (DS), stemming from the triplication of human chromosome 21, results in intellectual disability, with early mid-life onset of Alzheimer’s disease (AD) pathology. Early interventions to reduce cognitive impairments and neuropathology are lacking. One modality, maternal choline supplementation (MCS), has shown beneficial effects on behavior and gene expression in neurodevelopmental and neurodegenerative disorders, including trisomic mice. Loss of basal forebrain cholinergic neurons (BFCNs) and other DS/AD relevant hallmarks were observed in a well-established trisomic model (Ts65Dn, Ts). MCS attenuates these endophenotypes with beneficial behavioral effects in trisomic offspring. We postulate MCS ameliorates dysregulated cellular mechanisms within vulnerable BFCNs, with attenuation driven by novel gene expression. Here, choline acetyltransferase immunohistochemical labeling identified BFCNs in the medial septal/ventral diagonal band nuclei of the basal forebrain in Ts and normal disomic (2N) offspring at ~11 months of age from dams exposed to MCS or normal choline during the perinatal period. BFCNs (~500 per mouse) were microisolated and processed for RNA-sequencing. Bioinformatic assessment elucidated differentially expressed genes (DEGs) and pathway alterations in the context of genotype (Ts, 2N) and maternal diet (MCS, normal choline). MCS attenuated select dysregulated DEGs and relevant pathways in aged BFCNs. Trisomic MCS-responsive improvements included pathways such as cognitive impairment and nicotinamide adenine dinucleotide signaling, among others, indicative of increased behavioral and bioenergetic fitness. Although MCS does not eliminate the DS/AD phenotype, early choline delivery provides long-lasting benefits to aged trisomic BFCNs, indicating that MCS prolongs neuronal health in the context of DS/AD. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

9 pages, 477 KB  
Opinion
Underlying Piezo2 Channelopathy-Induced Neural Switch of COVID-19 Infection
by Balázs Sonkodi
Cells 2025, 14(15), 1182; https://doi.org/10.3390/cells14151182 - 31 Jul 2025
Viewed by 864
Abstract
The focal “hot spot” neuropathologies in COVID-19 infection are revealing footprints of a hidden underlying collapse of a novel ultrafast ultradian Piezo2 signaling system within the nervous system. Paradoxically, the same initiating pathophysiology may underpin the systemic findings in COVID-19 infection, namely the [...] Read more.
The focal “hot spot” neuropathologies in COVID-19 infection are revealing footprints of a hidden underlying collapse of a novel ultrafast ultradian Piezo2 signaling system within the nervous system. Paradoxically, the same initiating pathophysiology may underpin the systemic findings in COVID-19 infection, namely the multiorgan SARS-CoV-2 infection-induced vascular pathologies and brain–body-wide systemic pro-inflammatory signaling, depending on the concentration and exposure to infecting SARS-CoV-2 viruses. This common initiating microdamage is suggested to be the primary damage or the acquired channelopathy of the Piezo2 ion channel, leading to a principal gateway to pathophysiology. This Piezo2 channelopathy-induced neural switch could not only explain the initiation of disrupted cell–cell interactions, metabolic failure, microglial dysfunction, mitochondrial injury, glutamatergic synapse loss, inflammation and neurological states with the central involvement of the hippocampus and the medulla, but also the initiating pathophysiology without SARS-CoV-2 viral intracellular entry into neurons as well. Therefore, the impairment of the proposed Piezo2-induced quantum mechanical free-energy-stimulated ultrafast proton-coupled tunneling seems to be the principal and critical underlying COVID-19 infection-induced primary damage along the brain axes, depending on the loci of SARS-CoV-2 viral infection and intracellular entry. Moreover, this initiating Piezo2 channelopathy may also explain resultant autonomic dysregulation involving the medulla, hippocampus and heart rate regulation, not to mention sleep disturbance with altered rapid eye movement sleep and cognitive deficit in the short term, and even as a consequence of long COVID. The current opinion piece aims to promote future angles of science and research in order to further elucidate the not entirely known initiating pathophysiology of SARS-CoV-2 infection. Full article
(This article belongs to the Special Issue Insights into the Pathophysiology of NeuroCOVID: Current Topics)
Show Figures

Figure 1

17 pages, 937 KB  
Review
P-tau217 as a Biomarker in Alzheimer’s Disease: Applications in Latin American Populations
by Christian Alexis Varela-Vidales, Alejandra Martínez-Hernández, Elizabeth Hernández-Castellanos and Daniela L. C. Delgado-Lara
Int. J. Mol. Sci. 2025, 26(14), 6633; https://doi.org/10.3390/ijms26146633 - 10 Jul 2025
Viewed by 3603
Abstract
Alzheimer’s disease (AD) is one of the primary dementia causes worldwide. For this reason, there is a need for plasma-based diagnostic biomarkers to facilitate the timely diagnosis of AD. This work synthesizes the current evidence concerning the tau protein p-tau phosphorylated at threonine [...] Read more.
Alzheimer’s disease (AD) is one of the primary dementia causes worldwide. For this reason, there is a need for plasma-based diagnostic biomarkers to facilitate the timely diagnosis of AD. This work synthesizes the current evidence concerning the tau protein p-tau phosphorylated at threonine 217 (p-tau217) as an emerging biomarker, emphasizing its utility in preclinical phases and its potential application in Latin American populations. The findings indicate that p-tau217 has superior sensitivity and specificity compared to classical biomarkers such as p-tau181 and Aβ42. Likewise, its plasma concentration regulates neuropathological progression, as studies by Braak have shown, enabling it to identify alterations from the early stages. In Latin America, studies in Peru, Colombia, and Brazil have shown promising results, albeit with methodological limitations. Some of them have small sample sizes or lack neuroimaging confirmation. Additionally, clinical factors common in the region, such as hypertension, diabetes, or chronic kidney disease, may alter the clinical interpretation. In short, p-tau217 represents a potential non-invasive diagnostic resource. More diverse cohorts are needed to confirm its validity in daily clinical practice. Full article
Show Figures

Figure 1

13 pages, 1483 KB  
Article
Alzheimer’s Disease Lipidome: Elevated Cortical Levels of Glycerolipids in Subjects with Mild Cognitive Impairment (MCI) but Not in Non-Demented Alzheimer’s Neuropathology (NDAN) Subjects
by Paul L. Wood, John E. Cebak and Aaron W. Beger
J. Dement. Alzheimer's Dis. 2025, 2(3), 20; https://doi.org/10.3390/jdad2030020 - 1 Jul 2025
Viewed by 604
Abstract
Background/Objectives: Abnormal brain glycerolipid metabolism has been reported for Alzheimer’s disease (AD). This includes both diacylglycerols (DGs) and monogalactosyl-DGs (MGDGs), which are elevated in AD subjects. While DGs are also elevated in subjects with mild cognitive impairment (MCI), MGDGs have not yet [...] Read more.
Background/Objectives: Abnormal brain glycerolipid metabolism has been reported for Alzheimer’s disease (AD). This includes both diacylglycerols (DGs) and monogalactosyl-DGs (MGDGs), which are elevated in AD subjects. While DGs are also elevated in subjects with mild cognitive impairment (MCI), MGDGs have not yet been examined at this early stage of cognitive impairment. Methods: MGDG, triacylglycerol (TG), and ether glycerolipid levels in the cerebral cortex gray matter of controls, MCI, and non-demented Alzheimer’s neuropathology (NDAN) subjects were monitored by high-resolution mass spectrometry (<2 ppm mass error). Results: MGDG, MGDG ether, DG ether, and TG levels were elevated in the cerebral cortex of MCI but not NDAN subjects. Conclusions: A diverse array of glycerolipids was elevated in MCI subjects, suggesting a role in cognitive dysfunction. This suggestion is further supported by the maintenance of normal glycerolipid levels in NDAN subjects with amyloid accumulation but not cognitive deficits. Our data clearly indicate that while complex lipid alterations occur in MCI subjects, relative to controls 20 years younger, no such lipid alterations occur in NDAN subjects. While amyloid deposition in MCI is not involved in the observed lipid alterations, other ongoing neuropathologies may contribute to changes in lipid dynamics and vice versa. Full article
Show Figures

Figure 1

21 pages, 3636 KB  
Article
Antioxidant System Disturbances, Bioenergetic Disruption, and Glial Reactivity Induced by Methylmalonic Acid in the Developing Rat Brain
by Cristiano Antonio Dalpizolo, Josyane de Andrade Silveira, Manuela Bianchin Marcuzzo, Vitor Gayger-Dias, Vanessa-Fernanda Da Silva, Camila Vieira Pinheiro, Bruno Pereira dos Santos, Tiago Franco de Oliveira, Carlos-Alberto Gonçalves and Guilhian Leipnitz
Neuroglia 2025, 6(3), 25; https://doi.org/10.3390/neuroglia6030025 - 30 Jun 2025
Viewed by 1006
Abstract
Background: Elevated levels of methylmalonic acid (MMA) are observed in the bodily fluids and tissues of patients with methylmalonic aciduria, a metabolic disorder characterized by manifestations such as vomiting, lethargy, muscle weakness, seizures, and coma. Objectives and Methods: To better understand the neuropathological [...] Read more.
Background: Elevated levels of methylmalonic acid (MMA) are observed in the bodily fluids and tissues of patients with methylmalonic aciduria, a metabolic disorder characterized by manifestations such as vomiting, lethargy, muscle weakness, seizures, and coma. Objectives and Methods: To better understand the neuropathological mechanisms underlying this condition, we investigated the effects of intraperitoneal (i.p.) and intracerebroventricular (i.c.v.) administration of MMA on antioxidant defenses, citric acid cycle functioning, and glial reactivity in the cerebral cortex and striatum of Wistar rats. Amino acid levels were also quantified. Results: i.p. and i.c.v. administration of MMA decreased reduced glutathione levels and altered the activities of different antioxidant enzymes in the cortex and striatum. The activity of the citric acid cycle enzyme succinate dehydrogenase was diminished in both brain regions by i.p. and i.c.v. administration. Citrate synthase, isocitrate dehydrogenase, and malate dehydrogenase activities were further inhibited in the striatum. Furthermore, the i.p. administration increased glial fibrillary acidic protein (GFAP) and glucose transporter 1 (GLUT1) levels, whereas i.c.v. administration elevated GFAP and ionized calcium-binding adaptor molecule 1 (IBA1) levels in the striatum, suggesting glial activation. In contrast, no significant changes in glial markers were detected in the cortex. Moreover, synaptophysin levels remained unaltered in both regions. Finally, i.p. administration increased glutamate, glycine, and serine levels and reduced tyrosine concentrations in the striatum. Conclusions: Our findings indicate that oxidative stress, bioenergetic dysfunction, and glial reactivity induced by MMA may contribute to the neurological deficits observed in methylmalonic aciduria. Full article
Show Figures

Figure 1

29 pages, 1086 KB  
Review
Brain Neurotrophins and Plant Polyphenols: A Powerful Connection
by Marco Fiore, Sergio Terracina and Giampiero Ferraguti
Molecules 2025, 30(12), 2657; https://doi.org/10.3390/molecules30122657 - 19 Jun 2025
Cited by 4 | Viewed by 4577
Abstract
Neurodegenerative disorders, mental conditions, and cognitive decline represent significant challenges worldwide, with growing pieces of evidence implicating alterations in neurotrophin signaling as central to these diseases. Neurotrophins—such as nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF)—are indispensable for neuronal survival, differentiation, and [...] Read more.
Neurodegenerative disorders, mental conditions, and cognitive decline represent significant challenges worldwide, with growing pieces of evidence implicating alterations in neurotrophin signaling as central to these diseases. Neurotrophins—such as nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF)—are indispensable for neuronal survival, differentiation, and synaptic plasticity, and their dysregulation is closely associated with various neuropathological situations. Similarly, dietary plant polyphenols, abundant in vegetables, fruits, wine, tea, and extra virgin olive oil, show powerful anti-inflammatory, antioxidant, and anti-apoptotic activities. This narrative review critically addresses the evolving body of evidence that links plant polyphenols and brain neurotrophins, emphasizing several molecular mechanisms by which polyphenols regulate and modulate neurotrophin signaling. Crucial pathways include mitigation of neuroinflammatory responses, activation of intracellular cascades such as the cAMP response element-binding protein (CREB), epigenetic modulation, and the diminution of oxidative stress. Together, these effects contribute to potentiated enhanced synaptic function, neuronal integrity, and better learning and memory processes. Moreover, this narrative review examines how polyphenol-induced upregulation of neurotrophins may alleviate conditions associated not only with neurodegeneration but also with addiction and mood disorders, suggesting extensive therapeutic approaches. Findings from clinical investigations and animal models are presented to sustain the neuroprotective role of polyphenol-rich diets. Lastly, future research directions are recommended, focusing on polyphenol bioavailability optimization, considering combinatory dietary stratagems, and proposing personalized nutritional interventions. This wide-ranging perspective highlights plant polyphenols as encouraging modulators of neurotrophin pathways and supports their inclusion in approaches aimed at promoting brain health and counteracting neurodegenerative decline. Full article
(This article belongs to the Special Issue Exploring the Natural Antioxidants in Foods)
Show Figures

Graphical abstract

16 pages, 649 KB  
Review
Time-Frequency Domain Analysis of Quantitative Electroencephalography as a Biomarker for Dementia
by Chanda Simfukwe, Seong Soo A. An and Young Chul Youn
Diagnostics 2025, 15(12), 1509; https://doi.org/10.3390/diagnostics15121509 - 13 Jun 2025
Cited by 2 | Viewed by 1677
Abstract
Biomarkers currently used to diagnose dementia, including Alzheimer’s disease (AD), primarily detect molecular and structural brain changes associated with the condition’s pathology. Although these markers are pivotal in detecting disease-specific neuropathological hallmarks, their association with the clinical manifestations of dementia frequently remains poorly [...] Read more.
Biomarkers currently used to diagnose dementia, including Alzheimer’s disease (AD), primarily detect molecular and structural brain changes associated with the condition’s pathology. Although these markers are pivotal in detecting disease-specific neuropathological hallmarks, their association with the clinical manifestations of dementia frequently remains poorly defined and exhibits considerable variability. These biomarkers may show abnormalities in cognitively healthy individuals and frequently fail to accurately represent the severity of cognitive and functional impairments in individuals with dementia. Research indicates that synaptic degeneration and functional impairment occur early in the progression of AD and exhibit the strongest correlation with clinical symptoms. This identifies brain functional impairment measurements as promising early indicators for AD detection. Electroencephalography (EEG), a non-invasive and cost-effective method with high temporal resolution, is used as a biomarker for the early detection and diagnosis of AD through frequency-domain analysis of quantitative EEG (qEEG). Many researchers demonstrate that qEEG measures effectively identify disruptions in neuronal activity, including alterations in activity patterns, topographical distribution, and synchronization. Specific findings along the stages of AD include impaired neuronal synchronization, generalized EEG slowing, and an increase in lower-frequency bands accompanied by a decrease in higher-frequency bands of resting state EEG. Moreover, qEEG helps clinicians effectively correlate indicators of AD neuropathology and distinguish between various forms of dementia, positioning it as a promising, low-cost, non-invasive biomarker for dementia. However, additional clinical investigation is required to clarify the diagnostic and prognostic significance of qEEG measurements as early functional markers for AD. This narrative review examines time-frequency domain qEEG analysis as a potential biomarker across various types of dementia. Through a structured search of PubMed and Scopus, we identified studies assessing spectral and connectivity-based qEEG features. Consistent findings include EEG slowing, reduced functional connectivity, and network desynchronization. The review outlines key methodological challenges, such as lack of standardization and limited longitudinal validation, and recommends integrative, multimodal approaches to enhance diagnostic precision and clinical applicability. Full article
(This article belongs to the Section Clinical Diagnosis and Prognosis)
Show Figures

Figure 1

25 pages, 1948 KB  
Review
The Role and Pathogenesis of Tau Protein in Alzheimer’s Disease
by Xiaoyue Hong, Linshu Huang, Fang Lei, Tian Li, Yi Luo, Mengliu Zeng and Zhuo Wang
Biomolecules 2025, 15(6), 824; https://doi.org/10.3390/biom15060824 - 5 Jun 2025
Cited by 5 | Viewed by 5684
Abstract
Alzheimer’s disease (AD), a predominant neurodegenerative disorder, is clinically characterized by progressive cognitive deterioration and behavioral deficits. An in-depth understanding of the pathogenesis and neuropathology of AD is essential for the development of effective treatments and early diagnosis techniques. The neuropathological signature of [...] Read more.
Alzheimer’s disease (AD), a predominant neurodegenerative disorder, is clinically characterized by progressive cognitive deterioration and behavioral deficits. An in-depth understanding of the pathogenesis and neuropathology of AD is essential for the development of effective treatments and early diagnosis techniques. The neuropathological signature of AD involves two hallmark lesions: intraneuronal neurofibrillary tangles composed of hyperphosphorylated tau aggregates and extracellular senile plaques containing amyloid-β (Aβ) peptide depositions. Although Aβ-centric research has dominated AD investigations over the past three decades, pharmacological interventions targeting Aβ pathology have failed to demonstrate clinical efficacy. Tau, a microtubule-associated protein predominantly localized to neuronal axons, orchestrates microtubule stabilization and axonal transport through dynamic tubulin interactions under physiological conditions. In AD pathogenesis, however, tau undergoes pathogenic post-translational modifications (PTMs), encompassing hyperphosphorylation, lysine acetylation, methylation, ubiquitination, and glycosylation. These PTM-driven alterations induce microtubule network disintegration, mitochondrial dysfunction, synaptic impairment, and neuroinflammatory cascades, ultimately culminating in irreversible neurodegeneration and progressive cognitive decline. This review synthesizes contemporary advances in tau PTM research and delineates their mechanistic contributions to AD pathogenesis, thereby establishing a framework for biomarker discovery, targeted therapeutic development, and precision medicine approaches in tauopathies. This review synthesizes contemporary advances in tau PTM research and delineates their mechanistic contributions to AD pathogenesis, thereby establishing a solid theoretical and experimental basis for the early diagnosis of neurodegenerative diseases, the discovery of therapeutic targets, and the development of novel therapeutic strategies. Full article
(This article belongs to the Special Issue Pathogenesis and Neuropathology of Alzheimer's Disease)
Show Figures

Figure 1

19 pages, 978 KB  
Hypothesis
The Possible Role of Postnatal Biphasic Dysregulation of IGF-1 Tone in the Etiology of Idiopathic Autism Spectrum Disorder
by András Visegrády
Int. J. Mol. Sci. 2025, 26(10), 4483; https://doi.org/10.3390/ijms26104483 - 8 May 2025
Viewed by 2233
Abstract
Autism spectrum disorder (ASD) is a pervasive condition of neurodevelopmental origin with an increasing burden on society. Idiopathic ASD is notorious for its heterogeneous behavioral manifestations, and despite substantial efforts, its etiopathology is still unclear. An increasing amount of data points to the [...] Read more.
Autism spectrum disorder (ASD) is a pervasive condition of neurodevelopmental origin with an increasing burden on society. Idiopathic ASD is notorious for its heterogeneous behavioral manifestations, and despite substantial efforts, its etiopathology is still unclear. An increasing amount of data points to the causative role of critical developmental alterations in the first year of life, although the contribution of fetal, environmental, and genetic factors cannot be clearly distinguished. This review attempts to propose a narrative starting from neuropathological findings in ASD, involving insulin-like growth factor 1 (IGF-1) as a key modulator and demonstrates how the most consistent gestational risk factors of ASD–maternal insulin resistance and fetal growth insufficiency–converge at the perinatal dysregulation of offspring anabolism in the critical period of early development. A unifying hypothesis is derived, stating that the co-occurrence of these gestational conditions leads to postnatal biphasic dysregulation of IGF-1 tone in the offspring, leading first to insulin-dependent accelerated development, then to subsequent arrest of growth and brain maturation in ASD as an etiologic process. This hypothesis is tested for its explanation of various widely reported risk factors and observations of idiopathic ASD, including early postnatal growth abnormalities, the pervasive spectrum of symptoms, familial predisposition, and male susceptibility. Finally, further directions of research are outlined. Full article
Show Figures

Graphical abstract

27 pages, 7308 KB  
Article
PF-06447475 Molecule Attenuates the Neuropathology of Familial Alzheimer’s and Coexistent Parkinson’s Disease Markers in PSEN1 I416T Dopaminergic-like Neurons
by Diana Alejandra Quintero-Espinosa, Carlos Velez-Pardo and Marlene Jimenez-Del-Rio
Molecules 2025, 30(9), 2034; https://doi.org/10.3390/molecules30092034 - 2 May 2025
Viewed by 1226
Abstract
Familial Alzheimer’s disease (FAD) is a complex multifactorial disorder clinically characterized by cognitive impairment and memory loss. Pathologically, FAD is characterized by intracellular accumulation of the protein fragment Aβ42 (iAβ), hyperphosphorylated microtubule-associated protein TAU (p-TAU), and extensive degeneration of basal forebrain cholinergic neurons [...] Read more.
Familial Alzheimer’s disease (FAD) is a complex multifactorial disorder clinically characterized by cognitive impairment and memory loss. Pathologically, FAD is characterized by intracellular accumulation of the protein fragment Aβ42 (iAβ), hyperphosphorylated microtubule-associated protein TAU (p-TAU), and extensive degeneration of basal forebrain cholinergic neurons of the nucleus basalis of Meynert (NbM) and the medial septal nucleus (MSN), mainly caused by mutations in the amyloid precursor protein (APP), presenilin 1 (PSEN1), and PSEN2 gene. Since the dopaminergic system may contribute to FAD symptoms, alterations in the nigro-hippocampal pathway may be associated with cognitive impairment in FAD. Interestingly, p-α-synuclein (p-α-Syn), Aβ, and p-TAU have been found to coexist in vulnerable regions of postmortem AD brains. However, the mechanism by which Aβ, p-TAU, and α-Syn coexist in DAergic neurons in AD brains has not been determined. We generated PSEN1 I416T dopaminergic-like neurons (DALNs) from I416T menstrual stromal cells (MenSCs) in NeuroForsk 2.0 medium for 7 days and then cultured them in minimal culture medium (MCm) for another 4 days. On day 11, DALNs were analyzed for molecular and pathological markers by flow cytometry and fluorescence microscopy. We found that mutant DALNs showed increased accumulation of iAβ as well as increased phosphorylation of TAU at S202/T205 compared to WT DALNs. Thus, mutant DALNs exhibited typical pathological hallmarks of Alzheimer’s disease. Furthermore, PSEN1 I416T DALNs showed concomitant signs of OS as evidenced by the appearance of oxidized sensor protein DJ-1 (i.e., DJ-1C106-SO3) and apoptotic markers TP53, pS63-c-JUN, PUMA, and cleavage caspase 3 (CC3). Notably, these DALNs exhibited PD-associated proteins such as intracellular accumulation of α-Syn (detected as aggregates of pS129-α-Syn) and phosphorylation of LRRK2 kinase at residue S935. In addition, mutant DALNs showed a 17.16- and 6.17-fold decrease in DA-induced Ca2+ flux, compared to WT DALNs. These observations suggest that iAβ and p-TAU, together with p-α-Syn, and p-LRRK2 kinase, may damage DAergic neurons and thereby contribute to the exacerbation of neuropathologic processes in FAD. Remarkably, the LRRK2 inhibitor PF-06447475 (PF-475) significantly reversed PSEN1 I416T-induced neuropathological markers in DAergic neurons. PF-465 inhibitor reduced iAβ, oxDJ-1C106-SO3, and p-TAU. In addition, this inhibitor reduced pS935-LRRK2, pS129-αSYN, pS63-c-JUN, and CC3. We conclude that the observed neuroprotective effects of PF-475 are due to direct inhibition of LRRK2 activity and that the LRRK2 protein is upstream of the molecular cascade of apoptosis and proteinopathy. Our results suggest that PF-475 is an effective neuroprotective agent against endogenous PSEN1 I416T-induced neurotoxicity in DALNs coexisting with Parkinson’s disease markers. Therefore, PF-475 may be of great therapeutic value in FAD. Full article
(This article belongs to the Special Issue Therapeutic Agents for Neurodegenerative Disorders—2nd Edition)
Show Figures

Figure 1

Back to TopTop