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
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (130)

Search Parameters:
Keywords = CNS neurotoxicity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 3768 KiB  
Article
MWB_Analyzer: An Automated Embedded System for Real-Time Quantitative Analysis of Morphine Withdrawal Behaviors in Rodents
by Moran Zhang, Qianqian Li, Shunhang Li, Binxian Sun, Zhuli Wu, Jinxuan Liu, Xingchao Geng and Fangyi Chen
Toxics 2025, 13(7), 586; https://doi.org/10.3390/toxics13070586 - 14 Jul 2025
Viewed by 432
Abstract
Background/Objectives: Substance use disorders, particularly opioid addiction, continue to pose a major global health and toxicological challenge. Morphine dependence represents a significant problem in both clinical practice and preclinical research, particularly in modeling the pharmacodynamics of withdrawal. Rodent models remain indispensable for investigating [...] Read more.
Background/Objectives: Substance use disorders, particularly opioid addiction, continue to pose a major global health and toxicological challenge. Morphine dependence represents a significant problem in both clinical practice and preclinical research, particularly in modeling the pharmacodynamics of withdrawal. Rodent models remain indispensable for investigating the neurotoxicological effects of chronic opioid exposure and withdrawal. However, conventional behavioral assessments rely on manual observation, limiting objectivity, reproducibility, and scalability—critical constraints in modern drug toxicity evaluation. This study introduces MWB_Analyzer, an automated and high-throughput system designed to quantitatively and objectively assess morphine withdrawal behaviors in rats. The goal is to enhance toxicological assessments of CNS-active substances through robust, scalable behavioral phenotyping. Methods: MWB_Analyzer integrates optimized multi-angle video capture, real-time signal processing, and machine learning-driven behavioral classification. An improved YOLO-based architecture was developed for the accurate detection and categorization of withdrawal-associated behaviors in video frames, while a parallel pipeline processed audio signals. The system incorporates behavior-specific duration thresholds to isolate pharmacologically and toxicologically relevant behavioral events. Experimental animals were assigned to high-dose, low-dose, and control groups. Withdrawal was induced and monitored under standardized toxicological protocols. Results: MWB_Analyzer achieved over 95% reduction in redundant frame processing, markedly improving computational efficiency. It demonstrated high classification accuracy: >94% for video-based behaviors (93% on edge devices) and >92% for audio-based events. The use of behavioral thresholds enabled sensitive differentiation between dosage groups, revealing clear dose–response relationships and supporting its application in neuropharmacological and neurotoxicological profiling. Conclusions: MWB_Analyzer offers a robust, reproducible, and objective platform for the automated evaluation of opioid withdrawal syndromes in rodent models. It enhances throughput, precision, and standardization in addiction research. Importantly, this tool supports toxicological investigations of CNS drug effects, preclinical pharmacokinetic and pharmacodynamic evaluations, drug safety profiling, and regulatory assessment of novel opioid and CNS-active therapeutics. Full article
(This article belongs to the Section Drugs Toxicity)
Show Figures

Graphical abstract

18 pages, 306 KiB  
Review
The Interplay Between Melatonin and Nitric Oxide: Mechanisms and Implications in Stroke Pathophysiology
by Santos Blanco, María del Mar Muñoz-Gallardo, Raquel Hernández and María Ángeles Peinado
Antioxidants 2025, 14(6), 724; https://doi.org/10.3390/antiox14060724 - 13 Jun 2025
Viewed by 605
Abstract
This work reviews the complex interplay between melatonin and nitric oxide (NO) in the central nervous system (CNS), with a detailed focus on its involvement in stroke pathophysiology. Melatonin, a neurohormone with potent antioxidant, anti-inflammatory, and neuroprotective properties, and NO, a gaseous signaling [...] Read more.
This work reviews the complex interplay between melatonin and nitric oxide (NO) in the central nervous system (CNS), with a detailed focus on its involvement in stroke pathophysiology. Melatonin, a neurohormone with potent antioxidant, anti-inflammatory, and neuroprotective properties, and NO, a gaseous signaling molecule with diverse roles, interact crucially. In the context of ischemic stroke, NO exhibits a dual role: it can be neuroprotective (primarily via endothelial nitric oxide synthase (eNOS)) or neurotoxic (especially through inducible nitric oxide synthase (iNOS) and neuronal nitric oxide synthase (nNOS), contributing to the formation of damaging peroxynitrite (ONOO)). Melatonin has consistently demonstrated neuroprotective effects in animal models of stroke. Its key mechanisms related to NO include (1) differential modulation of nitric oxide synthase isoforms, suppressing detrimental iNOS expression/activity while often preserving or enhancing beneficial eNOS; (2) direct scavenging of NO and, critically, highly reactive peroxynitrite, thereby attenuating nitrosative stress; (3) reduction in neuroinflammation, partly by promoting M2 (anti-inflammatory) microglia polarization; and (4) mitochondrial protection and decreased apoptosis. These multifaceted actions of melatonin contribute to reduced infarct volume and improved functional outcomes, underscoring its considerable therapeutic potential for ischemic stroke through the favorable modulation of the melatonin–NO axis. Full article
17 pages, 2381 KiB  
Review
The Genetic Fingerprint of HIV in the Brain: Insights into Neurocognitive Dysfunction
by Sushama Jadhav, Shreeya Nair and Vijay Nema
Neuroglia 2025, 6(2), 23; https://doi.org/10.3390/neuroglia6020023 - 9 Jun 2025
Viewed by 737
Abstract
HIV, primarily targeting CD4 cells, infiltrates the CNS through various mechanisms, including chemokine-mediated signaling and blood–brain barrier disruption, leading to neuroinflammation and neuronal dysfunction. Viral proteins such as gp120, Tat, and Vpr directly induce neurotoxicity, oxidative stress, and mitochondrial dysfunction, exacerbating cognitive deficits [...] Read more.
HIV, primarily targeting CD4 cells, infiltrates the CNS through various mechanisms, including chemokine-mediated signaling and blood–brain barrier disruption, leading to neuroinflammation and neuronal dysfunction. Viral proteins such as gp120, Tat, and Vpr directly induce neurotoxicity, oxidative stress, and mitochondrial dysfunction, exacerbating cognitive deficits and motor impairments observed in HIV-associated neurocognitive disorders (HANDs). Host genetic factors, including CCR5 mutations and HLA alleles, influence susceptibility to HIV-related neurologic complications, shaping disease progression and treatment responses. Advanced molecular and bioinformatics techniques, from genome sequencing to structural modeling and network analysis, provide insights into viral pathogenesis and identify potential therapeutic targets. These findings underscore the future potential of precision medicine approaches tailored to individual genetic profiles to mitigate neurologic complications and improve outcomes in HIV-infected populations. This comprehensive review explores the intricate interplay between HIV infection and neurogenetics, focusing on how the virus impacts the central nervous system (CNS) and contributes to neurocognitive disorders. This report delves into how the virus influences genetic expression, neuroinflammation, and neurodegeneration, offering insights into molecular mechanisms behind HAND. Full article
Show Figures

Figure 1

25 pages, 1595 KiB  
Review
The Molecular Mechanisms of Cognitive Dysfunction in Long COVID: A Narrative Review
by Elena Popa, Andrei Emilian Popa, Mihaela Poroch, Vladimir Poroch, Monica Iuliana Ungureanu, Ana Maria Slanina, Agnes Bacusca and Elena Adorata Coman
Int. J. Mol. Sci. 2025, 26(11), 5102; https://doi.org/10.3390/ijms26115102 - 26 May 2025
Viewed by 3239
Abstract
Cognitive dysfunction represents one of the most persistent and disabling features of Long COVID, yet its molecular underpinnings remain incompletely understood. This narrative review synthesizes current evidence on the pathophysiological mechanisms linking SARS-CoV-2 infection to long-term neurocognitive sequelae. Key processes include persistent neuroinflammation, [...] Read more.
Cognitive dysfunction represents one of the most persistent and disabling features of Long COVID, yet its molecular underpinnings remain incompletely understood. This narrative review synthesizes current evidence on the pathophysiological mechanisms linking SARS-CoV-2 infection to long-term neurocognitive sequelae. Key processes include persistent neuroinflammation, blood–brain barrier (BBB) disruption, endothelial dysfunction, immune dysregulation, and neuroendocrine imbalance. Microglial activation and cytokine release (e.g., IL-6, TNF-α) promote synaptic dysfunction and neuronal injury, while activation of inflammasomes such as NLRP3 amplifies CNS inflammation. Vascular abnormalities, including microthrombosis and BBB leakage, facilitate the infiltration of peripheral immune cells and neurotoxic mediators. Hypothalamic–pituitary–adrenal axis dysfunction and reduced vagal tone further exacerbate systemic inflammation and autonomic imbalance. Biomarkers such as GFAP, NFL, IL-6, and S100B have been associated with both neuroinflammation and cognitive symptoms. Notably, transcriptomic signatures in Long COVID overlap with those observed in Alzheimer’s disease, highlighting shared pathways involving tau dysregulation, oxidative stress, and glial reactivity. Understanding these mechanisms is critical for identifying at-risk individuals and developing targeted therapeutic strategies. This review underscores the need for longitudinal research and integrative biomarker analysis to elucidate the molecular trajectory of cognitive impairment in Long COVID. Full article
Show Figures

Figure 1

23 pages, 741 KiB  
Review
Kynurenines and Mitochondrial Disturbances in Multiple Sclerosis
by Daniel Pukoli and László Vécsei
Int. J. Mol. Sci. 2025, 26(11), 5098; https://doi.org/10.3390/ijms26115098 - 26 May 2025
Cited by 1 | Viewed by 822
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease characterised by inflammation, demyelination, and neurodegeneration within the central nervous system. The pathogenesis of MS involves an immune-mediated attack on myelin and neurons, accompanied by blood–brain barrier dysfunction and chronic CNS inflammation. Central to MS [...] Read more.
Multiple sclerosis (MS) is a chronic autoimmune disease characterised by inflammation, demyelination, and neurodegeneration within the central nervous system. The pathogenesis of MS involves an immune-mediated attack on myelin and neurons, accompanied by blood–brain barrier dysfunction and chronic CNS inflammation. Central to MS pathology is dysregulation of the kynurenine pathway, which metabolises tryptophan into neuroactive compounds. Kynurenine pathway (KP) activation, driven by inflammatory cytokines, leads to the production of both neuroprotective (e.g., kynurenic acid, KYNA) and neurotoxic (e.g., quinolinic acid, QUIN) metabolites. Imbalance between these metabolites, particularly increased QUIN production, exacerbates glutamate excitotoxicity, oxidative stress, and mitochondrial dysfunction, contributing to neuronal and oligodendrocyte damage. Mitochondrial dysfunction plays a critical role in the pathophysiology of MS, exacerbating neurodegeneration through impaired energy metabolism and oxidative stress. This review integrates the current understanding of KP dysregulation in multiple sclerosis across disease stages. In RRMS, heightened KP activity correlates with inflammation and neuroprotection attempts through increased KYNA production. In contrast, SPMS and PPMS are associated with a shift towards a more neurotoxic KP profile, marked by elevated QUIN levels and reduced KYNA, exacerbating neurodegeneration and disability progression. Understanding these mechanisms offers insights into potential biomarkers and therapeutic targets for MS, emphasising the need for strategies to rebalance KP metabolism and mitigate neurotoxicity in progressive disease stages. Full article
Show Figures

Figure 1

37 pages, 1483 KiB  
Review
The Synergistic Roles of Glial Cells and Non-Coding RNAs in the Pathogenesis of Alzheimer’s Disease and Related Dementias (ADRDs)
by Sydney J. Risen, Devin Wahl, Thomas J. LaRocca and Julie A. Moreno
Neuroglia 2025, 6(2), 22; https://doi.org/10.3390/neuroglia6020022 - 6 May 2025
Viewed by 1583
Abstract
This review synthesizes the emerging understanding of the roles of glial cells and non-coding RNAs (ncRNAs) in the pathogenesis and progression of Alzheimer’s disease and related dementias (ADRDs). ADRDs encompass a spectrum of neurodegenerative disorders characterized by cognitive decline, memory impairment, and functional [...] Read more.
This review synthesizes the emerging understanding of the roles of glial cells and non-coding RNAs (ncRNAs) in the pathogenesis and progression of Alzheimer’s disease and related dementias (ADRDs). ADRDs encompass a spectrum of neurodegenerative disorders characterized by cognitive decline, memory impairment, and functional deterioration. The interplay between the most common types of glial cells—astrocytes, microglia, and oligodendrocytes—and ncRNAs is emerging as a critical factor in the development of ADRDs. Glial cells are essential for maintaining homeostasis within the central nervous system (CNS); however, their dysregulation can lead to neuroinflammation and neuronal dysfunction, exacerbating neurodegeneration. Reactive astrocytes and activated microglia can create neurotoxic environments that further impair neuronal health. Concurrently, ncRNAs, particularly long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), have emerged as significant regulators of glial gene expression, influencing inflammatory responses and glial cell function. Understanding the complex interactions between glial cells and ncRNAs is crucial for developing targeted therapeutic strategies. By elucidating the mechanisms underlying their interactions, this review aims to highlight the critical importance of glial cells and ncRNAs in the context of neurodegenerative diseases, paving the way for innovative approaches to prevent and treat ADRDs. Ultimately, enhancing our understanding of these processes may lead to novel therapies and improved outcomes for individuals affected by these debilitating conditions. Full article
Show Figures

Figure 1

14 pages, 1626 KiB  
Article
Development of Communication and Language Skills in Children with Hematological–Oncological Disorders: Challenges and Perspectives
by Giusy Melcarne, Giulia Marangon, Roberta Maria Incardona, Anna Agostinelli, Silvia Montino, Silvia Sorbara, Alessandra Biffi and Marta Tremolada
Children 2025, 12(5), 574; https://doi.org/10.3390/children12050574 - 29 Apr 2025
Viewed by 501
Abstract
Children with onco-hematological diseases require intensive medical treatments that can affect various aspects of their development. In addition to the disease itself, what influences the course of development most are the neurotoxic effects of therapies and frequent hospitalizations, especially if they occur in [...] Read more.
Children with onco-hematological diseases require intensive medical treatments that can affect various aspects of their development. In addition to the disease itself, what influences the course of development most are the neurotoxic effects of therapies and frequent hospitalizations, especially if they occur in the first three years of the child’s life. Among these challenges there is the potential for language delay, a condition that can impact their communication abilities and overall development. Background/Objectives: The aim of this study is to examine communicative and linguistic development in a small group of young children diagnosed with different forms of leukemia, rhabdomyosarcoma, and CNS tumors, recruited through the Hematology–Oncology Clinic of the Department of Child and Woman Health (University of Padova). Methods: Child direct (Griffiths III, PinG, PCGO) and parent indirect assessments (PVB, ABAS-II, ASCB) were provided. Results: Griffiths communication subscale scores in children were mainly below average (55.6%), and 44.4% attested at the clinical level in ABAS-II, with the ability to understand being significantly higher than the production of words. However, the two levels of assertiveness–responsiveness obtained balance in 66.7% of cases, and using the Griffiths personal subscale, only 22.2% of children attested below average. Conclusions: Understanding and addressing children’s communication needs is crucial to improve the quality of life of these young patients and foster optimal communicative and linguistic development despite the obstacles they face in order to implement interventions designed specifically for this type of population and their respective families, if necessary. Full article
(This article belongs to the Section Pediatric Hematology & Oncology)
Show Figures

Figure 1

14 pages, 24413 KiB  
Review
Rosenfeld’s Staining: A Valuable Tool for In Vitro Assessment of Astrocyte and Microglia Morphology
by Alana Alves Farias, Ana Carla dos Santos Costa, Jéssica Teles Souza, Érica Novaes Soares, Cinthia Cristina de Oliveira Santos Costa, Ravena Pereira do Nascimento, Silvia Lima Costa, Victor Diogenes Amaral da Silva and Maria de Fátima Dias Costa
Neuroglia 2025, 6(2), 16; https://doi.org/10.3390/neuroglia6020016 - 3 Apr 2025
Viewed by 1556
Abstract
In homeostasis, the glial cells support pivotal functions, such as neuronal differentiation, neuroprotection, nutrition, drug metabolism, and immune response in the central nervous system (CNS). Among these cells, astrocytes and microglia have been highlighted due to their role in the pathogenesis of several [...] Read more.
In homeostasis, the glial cells support pivotal functions, such as neuronal differentiation, neuroprotection, nutrition, drug metabolism, and immune response in the central nervous system (CNS). Among these cells, astrocytes and microglia have been highlighted due to their role in the pathogenesis of several diseases or due to their role in the defense against several insults (ex., chemicals, and pathogens). In Vitro cytological analysis of astrocytes and microglia has contributed to the understanding of the role of morphological changes in glial cells associated with a neuroprotective or neurotoxic phenotype. Currently, the main tools used for the investigation of glial cell morphology in culture are phase contrast microscopy or immunolabeling/fluorescence microscopy. However, generally, phase contrast microscopy does not generate images with high resolution and therefore does not contribute to visualizing a single cell morphology in confluent cell cultures. On the other hand, immunolabeling requires high-cost consumable antibodies, epifluorescence microscope or confocal microscope, and presents critical steps during the procedure. Therefore, identifying a fast, reproducible, low-cost alternative method that allows the evaluation of glial morphology is essential, especially for neuroscientists from low-income countries. This article aims to revise the use of Rosenfeld’s staining, as an alternative low-cost and easy-to-reproduce method to analyze astrocytic and microglial morphology in culture. Additionally, it shows Rosenfeld’s staining as a valuable tool to analyze changes in neural cell morphology in toxicological studies. Full article
Show Figures

Figure 1

15 pages, 920 KiB  
Review
Metabolic Syndrome and Schizophrenia: Adding a Piece to the Interplay Between the Kynurenine Pathway and Inflammation
by Jacopo Sapienza, Giulia Agostoni, Federica Repaci, Marco Spangaro, Stefano Comai and Marta Bosia
Metabolites 2025, 15(3), 176; https://doi.org/10.3390/metabo15030176 - 5 Mar 2025
Viewed by 1248
Abstract
The biology of schizophrenia is highly complex and multifaceted. Numerous efforts have been made over the years to disentangle the heterogeneity of the disease, gradually leading to a more detailed understanding of its underlying pathogenic mechanisms. Two cardinal elements in the pathophysiology of [...] Read more.
The biology of schizophrenia is highly complex and multifaceted. Numerous efforts have been made over the years to disentangle the heterogeneity of the disease, gradually leading to a more detailed understanding of its underlying pathogenic mechanisms. Two cardinal elements in the pathophysiology of schizophrenia are neuroinflammation and alterations of neurotransmission. The kynurenine (KYN) pathway (KP) is of particular importance because it is inducted by systemic low-grade inflammation in peripheral tissues, producing metabolites that are neuroactive (i.e., modulating glutamatergic and cholinergic neurotransmission), neuroprotective, or neurotoxic. Consequently, the KP is at the crossroads between two primary systems involved in the pathogenesis of schizophrenia. It bridges the central nervous system (CNS) and the periphery, as KP metabolites can cross the blood–brain barrier and modulate neuronal activity. Metabolic syndrome plays a crucial role in this context, as it frequently co-occurs with schizophrenia, contributing to a sub-inflammatory state able to activate the KP. This narrative review provides valuable insights into these complex interactions, offering a framework for developing targeted therapeutic interventions or precision psychiatry approaches of the disorder. Full article
(This article belongs to the Special Issue The Interplay Between Inflammation and Metabolism in Disease)
Show Figures

Figure 1

11 pages, 1296 KiB  
Article
Concerning Mercury (Hg) Levels in the Hair of Children Inhabiting a Volcanically Active Area
by Rute Fontes, Nádia M. P. Coelho, Patrícia V. Garcia, Filipe Bernardo and Armindo S. Rodrigues
Toxics 2025, 13(3), 146; https://doi.org/10.3390/toxics13030146 - 21 Feb 2025
Viewed by 753
Abstract
Background: Gaseous elemental mercury (Hg0 or GEM) is an atmospheric form of mercury (Hg)—a toxic heavy metal—that is naturally released in volcanic environments. Research with wild mice demonstrates that chronic exposure to a hydrothermal volcanic environment leads to the bioaccumulation of Hg [...] Read more.
Background: Gaseous elemental mercury (Hg0 or GEM) is an atmospheric form of mercury (Hg)—a toxic heavy metal—that is naturally released in volcanic environments. Research with wild mice demonstrates that chronic exposure to a hydrothermal volcanic environment leads to the bioaccumulation of Hg in the lungs, but also in both the central (CNS) and peripheric (PNS) nervous systems, with marked indications of neurotoxicity. Studies addressing human exposure to volcanogenic Hg0 are scarce, hence its risks are still unknown. This study aims to evaluate the level of exposure to Hg0 in children living in a volcanically active environment. Methodology and main findings: Two groups of school-aged children (from 6 to 9 years old) were part of this study: one with children inhabiting a hydrothermal area (exposed group) and another with children inhabiting an area without volcanic activity (non-exposed group). Hair samples were collected from each individual for Hg level analysis. It was found that the levels of Hg in the hair of exposed children were 4.2 times higher than in that of non-exposed children (≈1797.84 ± 454.92 ppb vs. 430.69 ± 66.43 ppb, respectively). Conclusion: Given the vast health risks Hg poses, the need to monitor the health of populations inhabiting volcanically active areas is highlighted. Because little is known about the fate, modifications, and effects of Hg0 in the human body, particularly regarding its effects on the nervous system in children, the development of further research within the scope is strongly encouraged. Full article
Show Figures

Graphical abstract

23 pages, 1102 KiB  
Review
Botulinum Toxin: A Comprehensive Review of Its Molecular Architecture and Mechanistic Action
by Raj Kumar and Bal Ram Singh
Int. J. Mol. Sci. 2025, 26(2), 777; https://doi.org/10.3390/ijms26020777 - 17 Jan 2025
Cited by 5 | Viewed by 5931
Abstract
Botulinum toxin (BoNT), the most potent substance known to humans, likely evolved not to kill but to serve other biological purposes. While its use in cosmetic applications is well known, its medical utility has become increasingly significant due to the intricacies of its [...] Read more.
Botulinum toxin (BoNT), the most potent substance known to humans, likely evolved not to kill but to serve other biological purposes. While its use in cosmetic applications is well known, its medical utility has become increasingly significant due to the intricacies of its structure and function. The toxin’s structural complexity enables it to target specific cellular processes with remarkable precision, making it an invaluable tool in both basic and applied biomedical research. BoNT’s potency stems from its unique structural features, which include domains responsible for receptor recognition, membrane binding, internalization, and enzymatic cleavage. This division of labor within the toxin’s structure allows it to specifically recognize and interact with synaptic proteins, leading to precise cleavage at targeted sites within neurons. The toxin’s mechanism of action involves a multi-step process: recognition, binding, and catalysis, ultimately blocking neurotransmitter release by cleaving proteins like SNAP-25, VAMP, and syntaxin. This disruption in synaptic vesicle fusion causes paralysis, typically in peripheral neurons. However, emerging evidence suggests that BoNT also affects the central nervous system (CNS), influencing presynaptic functions and distant neuronal systems. The evolutionary history of BoNT reveals that its neurotoxic properties likely provided a selective advantage in certain ecological contexts. Interestingly, the very features that make BoNT a potent toxin also enable its therapeutic applications, offering precision in treating neurological disorders like dystonia, spasticity, and chronic pain. In this review, we highlight the toxin’s structural, functional, and evolutionary aspects, explore its clinical uses, and identify key research gaps, such as BoNT’s central effects and its long-term cellular impact. A clear understanding of these aspects could facilitate the representation of BoNT as a unique scientific paradigm for studying neuronal processes and developing targeted therapeutic strategies. Full article
(This article belongs to the Collection Feature Papers in Molecular Toxicology)
Show Figures

Figure 1

50 pages, 3786 KiB  
Review
The Crucial Role of the Blood–Brain Barrier in Neurodegenerative Diseases: Mechanisms of Disruption and Therapeutic Implications
by Sehwan Kim, Un Ju Jung and Sang Ryong Kim
J. Clin. Med. 2025, 14(2), 386; https://doi.org/10.3390/jcm14020386 - 9 Jan 2025
Cited by 8 | Viewed by 4430
Abstract
The blood–brain barrier (BBB) is a crucial structure that maintains brain homeostasis by regulating the entry of molecules and cells from the bloodstream into the central nervous system (CNS). Neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease, as well as ischemic stroke, compromise [...] Read more.
The blood–brain barrier (BBB) is a crucial structure that maintains brain homeostasis by regulating the entry of molecules and cells from the bloodstream into the central nervous system (CNS). Neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease, as well as ischemic stroke, compromise the integrity of the BBB. This leads to increased permeability and the infiltration of harmful substances, thereby accelerating neurodegeneration. In this review, we explore the mechanisms underlying BBB disruption, including oxidative stress, neuroinflammation, vascular dysfunction, and the loss of tight junction integrity, in patients with neurodegenerative diseases. We discuss how BBB breakdown contributes to neuroinflammation, neurotoxicity, and the abnormal accumulation of pathological proteins, all of which exacerbate neuronal damage and facilitate disease progression. Furthermore, we discuss potential therapeutic strategies aimed at preserving or restoring BBB function, such as anti-inflammatory treatments, antioxidant therapies, and approaches to enhance tight junction integrity. Given the central role of the BBB in neurodegeneration, maintaining its integrity represents a promising therapeutic approach to slow or prevent the progression of neurodegenerative diseases. Full article
(This article belongs to the Section Clinical Neurology)
Show Figures

Figure 1

21 pages, 7165 KiB  
Article
Evaluation of Hippocampal Microanatomy and Neuro-Biomarkers Following Administration of Silver Nanoparticles Conjugated with Tenofovir Disoproxil Fumarate in Experimental Diabetic Rats
by Sodiq Kolawole Lawal, Samuel Oluwaseun Olojede, Babatunde Adebola Alabi, Kafalotse Sylvia Dithole, Samuel Thopho Matula, Edwin Coleridge Naidu, Carmen Olivia Rennie and Onyemaechi Okpara Azu
Pharmaceuticals 2024, 17(12), 1635; https://doi.org/10.3390/ph17121635 - 5 Dec 2024
Viewed by 964
Abstract
Adverse complications like metabolic disorders, neurotoxicity, and low central nervous system (CNS) penetration are associated with the long-term use of tenofovir disoproxil fumarate (TDF). Therefore, some modifications are required to enhance neurological functions using silver nanoparticles (AgNPs). This study aimed to evaluate the [...] Read more.
Adverse complications like metabolic disorders, neurotoxicity, and low central nervous system (CNS) penetration are associated with the long-term use of tenofovir disoproxil fumarate (TDF). Therefore, some modifications are required to enhance neurological functions using silver nanoparticles (AgNPs). This study aimed to evaluate the neuroprotective impact of silver nanoparticles (AgNPs)-conjugated TDF as AgNPs-TDF on the hippocampal microanatomy and some neuro-biomarkers of diabetic rats. Forty-two male Sprague-Dawley rats, with an average weight of 250 ± 13 g, were divided into non-diabetic and diabetic groups. They were further divided into 3 groups each (n = 7): non-diabetic control (NC), non-diabetic + TDF (NTF), and non-diabetic + TDF + silver nanoparticles (NTS), as well as diabetic control (DC), diabetic + TDF (DTF), and diabetic + TDF + silver nanoparticles (DTS). The characterization of AgNPs-TDF was assessed, and the conjugates were administered to the diabetic rats, followed by behavioral testing and biochemical, immunohistochemical, and microanatomy analyses of the hippocampus. The results showed that the administration of AgNPs-TDF significantly reduced the blood glucose level, malondialdehyde (MDA), and inflammatory biomarker concentrations in DTS compared with the DTF and DC groups. Furthermore, AgNPs-TDF administration significantly increased the levels of tissue superoxide dismutase (SOD), reduced glutathione (GSH), and insulin-like growth factor-1 in DTS compared with the DTF and DC groups. In addition, the DTS group revealed a monomorphic pattern of dark-stained neuronal nuclei similar to the control group and showed neuroprotective effects on hippocampal microanatomy compared with the DTF group. This study shows that AgNPs-TDF restores various alterations in the hippocampus and improves cognitive functions in diabetic rats. Full article
(This article belongs to the Special Issue Therapeutic Potential of Silver Nanoparticles (AgNPs))
Show Figures

Figure 1

22 pages, 931 KiB  
Review
CAR-T Cells for the Treatment of Central Nervous System Tumours: Known and Emerging Neurotoxicities
by Leonardo Palazzo, Valentina Pieri, Giulia Berzero and Massimo Filippi
Brain Sci. 2024, 14(12), 1220; https://doi.org/10.3390/brainsci14121220 - 30 Nov 2024
Viewed by 2079
Abstract
The advent of chimeric antigen receptor (CAR)-T cells has recently changed the prognosis of relapsing/refractory diffuse large B-cell lymphomas, showing response rates as high as 60 to 80%. Common toxicities reported in the pivotal clinical trials include the cytokine release syndrome (CRS) and [...] Read more.
The advent of chimeric antigen receptor (CAR)-T cells has recently changed the prognosis of relapsing/refractory diffuse large B-cell lymphomas, showing response rates as high as 60 to 80%. Common toxicities reported in the pivotal clinical trials include the cytokine release syndrome (CRS) and the Immune effector Cell-Associated Neurotoxicity Syndrome (ICANS), a stereotyped encephalopathy related to myeloid cell activation and blood–brain barrier dysfunction, presenting with a distinctive cascade of dysgraphia, aphasia, disorientation, attention deficits, vigilance impairment, motor symptoms, seizures, and diffuse brain oedema. The tremendous oncological efficacy of CAR-T cells observed in systemic B-cell malignancies is leading to their growing use in patients with primary or secondary central nervous system (CNS) lymphomas and in patients with solid tumours, including several CNS cancers. Early studies conducted in adult and paediatric patients with solid CNS tumours reported a distinct profile of neurotoxicity referred to as Tumour inflammation-associated neurotoxicity (TIAN), corresponding to local inflammation at the tumour site manifesting with focal neurological deficits or mechanical complications (e.g., obstructive hydrocephalus). The present review summarises available data on the efficacy and safety of CAR-T cells for solid and haematological CNS malignancies, emphasising known and emerging phenotypes, ongoing challenges, and future perspectives. Full article
(This article belongs to the Special Issue Neurotoxicities from Cancer Immunotherapies)
Show Figures

Figure 1

34 pages, 3816 KiB  
Review
The Yin and Yang of Microglia-Derived Extracellular Vesicles in CNS Injury and Diseases
by Mousumi Ghosh and Damien D. Pearse
Cells 2024, 13(22), 1834; https://doi.org/10.3390/cells13221834 - 6 Nov 2024
Cited by 6 | Viewed by 4714
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in maintaining neural homeostasis but can also contribute to disease and injury when this state is disrupted or conversely play a pivotal role in neurorepair. One way that [...] Read more.
Microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in maintaining neural homeostasis but can also contribute to disease and injury when this state is disrupted or conversely play a pivotal role in neurorepair. One way that microglia exert their effects is through the secretion of small vesicles, microglia-derived exosomes (MGEVs). Exosomes facilitate intercellular communication through transported cargoes of proteins, lipids, RNA, and other bioactive molecules that can alter the behavior of the cells that internalize them. Under normal physiological conditions, MGEVs are essential to homeostasis, whereas the dysregulation of their production and/or alterations in their cargoes have been implicated in the pathogenesis of numerous neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), spinal cord injury (SCI), and traumatic brain injury (TBI). In contrast, MGEVs may also offer therapeutic potential by reversing inflammation or being amenable to engineering for the delivery of beneficial biologics or drugs. The effects of MGEVs are determined by the phenotypic state of the parent microglia. Exosomes from anti-inflammatory or pro-regenerative microglia support neurorepair and cell survival by delivering neurotrophic factors, anti-inflammatory mediators, and molecular chaperones. Further, MGEVs can also deliver components like mitochondrial DNA (mtDNA) and proteins to damaged neurons to enhance cellular metabolism and resilience. MGEVs derived from pro-inflammatory microglia can have detrimental effects on neural health. Their cargo often contains pro-inflammatory cytokines, molecules involved in oxidative stress, and neurotoxic proteins, which can exacerbate neuroinflammation, contribute to neuronal damage, and impair synaptic function, hindering neurorepair processes. The role of MGEVs in neurodegeneration and injury—whether beneficial or harmful—largely depends on how they modulate inflammation through the pro- and anti-inflammatory factors in their cargo, including cytokines and microRNAs. In addition, through the propagation of pathological proteins, such as amyloid-beta and alpha-synuclein, MGEVs can also contribute to disease progression in disorders such as AD and PD, or by the transfer of apoptotic or necrotic factors, they can induce neuron toxicity or trigger glial scarring during neurological injury. In this review, we have provided a comprehensive and up-to-date understanding of the molecular mechanisms underlying the multifaceted role of MGEVs in neurological injury and disease. In particular, the role that specific exosome cargoes play in various pathological conditions, either in disease progression or recovery, will be discussed. The therapeutic potential of MGEVs has been highlighted including potential engineering methodologies that have been employed to alter their cargoes or cell-selective targeting. Understanding the factors that influence the balance between beneficial and detrimental exosome signaling in the CNS is crucial for developing new therapeutic strategies for neurodegenerative diseases and neurotrauma. Full article
Show Figures

Figure 1

Back to TopTop