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

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
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,095)

Search Parameters:
Keywords = neuroprotective agents

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
33 pages, 885 KB  
Review
The Neuroprotective Potential of Ocimum Plant Species: Seasoning the Mind with Sweet and Holy Basil
by Alexandru Vasincu, Răzvan-Nicolae Rusu, Daniela-Carmen Ababei, Delia Bulea, Oana Dana Arcan, Ioana Mirela Vasincu, Sorin Beșchea Chiriac, Ionuț-Răducu Popescu, Walther Bild and Veronica Bild
Nutrients 2025, 17(17), 2877; https://doi.org/10.3390/nu17172877 - 5 Sep 2025
Abstract
Neurodegenerative diseases (NDs) encompass a group of chronic conditions, characterized by neuronal losses in large areas of the brain, leading to cognitive and behavioral impairments. Alzheimer’s Disease (AD), the most common form of dementia, is a progressive ND, characterized by the accumulation of [...] Read more.
Neurodegenerative diseases (NDs) encompass a group of chronic conditions, characterized by neuronal losses in large areas of the brain, leading to cognitive and behavioral impairments. Alzheimer’s Disease (AD), the most common form of dementia, is a progressive ND, characterized by the accumulation of amyloid β and tau protein, entails cognitive decline, neuroinflammation, mitochondrial dysfunction, and blood–brain barrier impairment, with oxidative stress playing a critical role in its pathogenesis. To date, the available pharmacotherapy has shown limited efficacy, and multitarget activity of plant-derived neuroprotective bioactive compounds is currently in focus. This review synthesizes experimental evidence regarding Ocimum species with neuroprotective potential in AD, particularly Ocimum sanctum and Ocimum basilicum. These plants are rich in bioactive compounds including polyphenols, flavonoids, essential oils, and triterpenoids that synergistically scavenge reactive oxygen/nitrogen species, upregulate endogenous antioxidant enzymes (SOD, CAT, and GPx), and reduce lipid peroxidation. Furthermore, these extracts have demonstrated the ability to decrease β-amyloid accumulation and tau protein levels, key pathological features of AD. Even though additional research is required to fully assess their potential as therapeutic agents for NDs, by diving into the specific mechanisms through which they improve neurodegenerative processes, important steps can be made towards this endpoint. Full article
(This article belongs to the Special Issue Therapeutic Potential of Phytochemicals in Neurodegenerative Diseases)
24 pages, 2790 KB  
Article
Anti-HMGB1 Antibody Therapy Ameliorates Spinal Cord Ischemia–Reperfusion Injury in Rabbits
by Genya Muraoka, Yasuhiro Fujii, Keyue Liu, Handong Qiao, Dengli Wang, Daiki Ousaka, Susumu Oozawa, Shingo Kasahara and Masahiro Nishibori
Int. J. Mol. Sci. 2025, 26(17), 8643; https://doi.org/10.3390/ijms26178643 - 5 Sep 2025
Viewed by 42
Abstract
Spinal cord ischemia–reperfusion (SCI/R) injury remains a major clinical challenge with limited therapeutic options. High-mobility group box 1 (HMGB1), a proinflammatory mediator released during cellular stress, has been implicated in the pathogenesis of ischemia–reperfusion-induced neural damage. In this study, we investigated the neuroprotective [...] Read more.
Spinal cord ischemia–reperfusion (SCI/R) injury remains a major clinical challenge with limited therapeutic options. High-mobility group box 1 (HMGB1), a proinflammatory mediator released during cellular stress, has been implicated in the pathogenesis of ischemia–reperfusion-induced neural damage. In this study, we investigated the neuroprotective potential of the anti-HMGB1 monoclonal antibody (mAb) in a rabbit model of SCI/R injury. Male New Zealand White rabbits were anesthetized and subjected to 11 min of abdominal aortic occlusion using a micro-bulldog clamp following heparinization. Anti-HMGB1 mAb or control IgG was administered intravenously immediately after reperfusion and again at 6 h post-reperfusion. Neurological function was assessed at 6, 24, and 48 h after reperfusion using the modified Tarlov scoring system. The rabbits were euthanized 48 h after reperfusion for spinal cord and blood sampling. Treatment with anti-HMGB1 mAb significantly improved neurological outcomes, reduced the extent of spinal cord infarction, preserved motor neuron viability, and decreased the presence of activated microglia and infiltrating neutrophils. Furthermore, it attenuated apoptosis, oxidative stress, and inflammatory responses in the spinal cord, and helped maintain the integrity of the blood–spinal cord barrier. These findings suggest that anti-HMGB1 mAb may serve as a promising therapeutic agent for SCI/R injury. Full article
(This article belongs to the Collection Advances in Cell and Molecular Biology)
Show Figures

Figure 1

31 pages, 6911 KB  
Review
Advances in Gold Nanoparticles for the Diagnosis and Management of Alzheimer’s Disease
by Bhagavathi Sundaram Sivamaruthi, Periyanaina Kesika, Natarajan Sisubalan and Chaiyavat Chaiyasut
Pharmaceutics 2025, 17(9), 1158; https://doi.org/10.3390/pharmaceutics17091158 - 3 Sep 2025
Viewed by 272
Abstract
Alzheimer’s disease (AD) presents a significant challenge in modern healthcare, prompting exploration into novel therapeutic strategies. This review clearly classifies different types of gold (Au) nanoparticles (NPs) (AuNPs), links them to the gut–brain axis, highlights recent advances, and points out future research needs, [...] Read more.
Alzheimer’s disease (AD) presents a significant challenge in modern healthcare, prompting exploration into novel therapeutic strategies. This review clearly classifies different types of gold (Au) nanoparticles (NPs) (AuNPs), links them to the gut–brain axis, highlights recent advances, and points out future research needs, offering a more updated perspective than earlier reviews. Diverse approaches have emerged from single to hybrid and functionalized AuNPs, including innovative nanotherapeutic agents like Au nanorods-polyethylene glycol-angiopep-2 peptide/D1 peptide and noninvasive dynamic magnetic field-stimulated NPs. AuNPs have been reported for the neuroprotective properties. Clinical applications of AuNPs highlight their promise in diagnosis and therapeutic monitoring. However, challenges persist, notably in overcoming blood–brain barrier limitations and refining drug delivery systems. Furthermore, the incomplete understanding of AD’s physiological and pathological mechanisms hinders therapeutic development. Future research directions should prioritize elucidating these mechanisms and optimizing AuNPs physicochemical properties for therapeutic efficacy. Despite limitations, nanomaterial-based therapies hold promise for revolutionizing AD treatment and addressing other central nervous system disorders. It also emphasizes the importance of further investigation into the potential of AuNPs, envisioning a future where they serve as a cornerstone in advancing neurological healthcare. Full article
Show Figures

Figure 1

16 pages, 4550 KB  
Article
Methylation Enables Sensitive LC-MS/MS Quantification of Ciclopirox in a Mouse Pharmacokinetics Study
by Roshan Katekar, Zhengqiang Wang and Jiashu Xie
Molecules 2025, 30(17), 3599; https://doi.org/10.3390/molecules30173599 - 3 Sep 2025
Viewed by 249
Abstract
Ciclopirox (CPX), a topical antifungal agent of the N-hydroxypyridone class, has gained renewed interest for its potential anticancer, antiviral, antibacterial, and neuroprotective effects. However, due to lack of reliable validated bioanalytical methods, current insights into its pharmacokinetics profile beyond topical use remain limited. [...] Read more.
Ciclopirox (CPX), a topical antifungal agent of the N-hydroxypyridone class, has gained renewed interest for its potential anticancer, antiviral, antibacterial, and neuroprotective effects. However, due to lack of reliable validated bioanalytical methods, current insights into its pharmacokinetics profile beyond topical use remain limited. To support therapeutic repurposing, we developed and validated a rapid, sensitive LC-MS/MS method for systemic pharmacokinetic evaluation in mice. The method employs methyl derivatization of CPX’s N-hydroxy group, producing methylated CPX (Me-CPX) for improved chromatographic performance which was subsequently retained on the AtlantisTM T3 C18 reverse phase column. Concentration of CPX is determined indirectly based on the measured response of Me-CPX. The method achieved excellent recovery, a 4-min rapid runtime, sensitivity with LLOQ of 3.906 nM (0.81 ng/mL), and a linear range up to 1000 nM (r ≥ 0.9998). All validation parameters including intra- and inter-day accuracy, precision, matrix effects, stability and dilution integrity met the criteria defined by regulatory International Council for Harmonisation (ICH) M10 bioanalytical method validation guidelines. Application of the method to in vitro plasma protein binding studies revealed high protein binding (>99%) of CPX in both human and mice plasma. Preliminary PK analysis following intravenous and oral administration in CD-1 mice demonstrated moderate systemic exposure after oral dosing, with an estimated absolute bioavailability of 52.5%. These findings establish the method’s suitability and robustness for preclinical and future clinical development of CPX as a repurposed therapeutic agent. Full article
Show Figures

Graphical abstract

37 pages, 2146 KB  
Review
The Central Nervous System Modulatory Activities of N-Acetylcysteine: A Synthesis of Two Decades of Evidence
by Desislava Ivanova Cherneva, Gabriela Kehayova, Simeonka Dimitrova and Stela Dragomanova
Curr. Issues Mol. Biol. 2025, 47(9), 710; https://doi.org/10.3390/cimb47090710 - 1 Sep 2025
Viewed by 360
Abstract
N-acetylcysteine (NAC) has garnered increasing interest for its neurotherapeutic capabilities beyond its recognized functions as a mucolytic agent and an antidote for acetaminophen toxicity. This review consolidates findings from both preclinical and clinical studies to investigate NAC’s diverse modulatory effects on the central [...] Read more.
N-acetylcysteine (NAC) has garnered increasing interest for its neurotherapeutic capabilities beyond its recognized functions as a mucolytic agent and an antidote for acetaminophen toxicity. This review consolidates findings from both preclinical and clinical studies to investigate NAC’s diverse modulatory effects on the central nervous system (CNS). NAC primarily functions as an antioxidant by replenishing glutathione and mitigating oxidative stress; however, it produces glutathione-independent effects through the modulation of mitochondrial redox systems, ferroptosis, and the Nrf2-ARE signaling pathway. It plays a significant role in neuroinflammatory processes by inhibiting the production of cytokines, the expression of iNOS, and the activation of microglia. Furthermore, NAC affects various neurotransmitter systems—including glutamatergic, dopaminergic, GABAergic, serotonergic, cholinergic, and adrenergic pathways—by modulating synaptic transmission, receptor activity, and transporter functionality. It promotes neuroprotection through the enhancement of neurotrophic factors, the preservation of mitochondrial integrity, and the upregulation of survival signaling pathways. Recent evidence also emphasizes NAC’s role in gene expression and the regulation of cortisol levels. The extensive range of NAC’s neurobiological effects highlights its therapeutic potential in treating neurodegenerative and neuropsychiatric disorders. Nevertheless, the variability in clinical outcomes indicates a pressing need for more focused, mechanism-based research. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Graphical abstract

30 pages, 1153 KB  
Review
A Review of the Mechanisms and Risks of Panax ginseng in the Treatment of Alcohol Use Disorder
by Eli Frazer, Candi Zhao, Jacky Lee, Jonathan Shaw, Charles Lai, Peter Bota and Tina Allee
Diseases 2025, 13(9), 285; https://doi.org/10.3390/diseases13090285 - 1 Sep 2025
Viewed by 371
Abstract
Alcohol use disorder (AUD) is a widespread, multifaceted disorder involving overproduction of pro-inflammatory cytokines, oxidative liver injury, and dysfunction of the brain’s dopaminergic reward circuits. Korean red ginseng (KRG), an herbal supplement derived from Panax ginseng, has demonstrated qualities potentially useful to [...] Read more.
Alcohol use disorder (AUD) is a widespread, multifaceted disorder involving overproduction of pro-inflammatory cytokines, oxidative liver injury, and dysfunction of the brain’s dopaminergic reward circuits. Korean red ginseng (KRG), an herbal supplement derived from Panax ginseng, has demonstrated qualities potentially useful to the treatment of AUD, including antioxidative, anti-inflammatory, neuroprotective, and anxiolytic effects. This review examines active constituents of KRG, their pharmacological actions, and evidence supporting KRG’s therapeutic potential in the context of AUD, while also assessing its safety profile, adverse effects, and potential drug interactions. KRG’s main bioactive constituents, ginsenosides, appear to have roles in modulating alcohol-metabolizing enzymes, ethanol-activated inflammatory cytokine cascades, and neurological systems disrupted by AUD, including GABAergic and dopaminergic pathways. Evidence from animal models and limited small-scale human trials suggests KRG may alleviate symptoms of alcohol withdrawal, enhance cognitive performance, and attenuate anxiety through these pathways. While generally safe for consumption, several case reports and animal studies have indicated KRG’s potential to pose a variety of risks in vulnerable populations at high, prolonged doses, including hepatotoxicity, cardiovascular changes, mood disturbances, and hormonal effects. Furthermore, KRG’s neuromodulating role and influence on cytochrome P450 enzymes make it liable to interact with several medications, including warfarin, midazolam, selegiline, and serotonergic agents. Overall, KRG shows promise as a complementary supplement in managing aspects of AUD, though current evidence is limited by low sample sizes, inconsistent reports regarding nuances of ginsenosides’ mechanisms, and a low number of human trials. Further human-focused research is needed to elucidate its safety, efficacy, and mechanism. Full article
(This article belongs to the Section Neuro-psychiatric Disorders)
Show Figures

Figure 1

27 pages, 1992 KB  
Review
Therapeutic Potential of Natural Compounds for Brain Ischemia-Reperfusion Injury
by Ki-Yeon Yoo, Moo-Ho Won, Ji Hyeon Ahn and Joon Ha Park
Biology 2025, 14(9), 1153; https://doi.org/10.3390/biology14091153 - 1 Sep 2025
Viewed by 352
Abstract
Brain ischemia-reperfusion (I/R) injury, commonly occurring in ischemic stroke and post-cardiac arrest scenarios, results in complex secondary damage involving oxidative stress, inflammation, apoptosis, and blood-brain barrier (BBB) breakdown. Despite decades of research, no pharmacological agent has yet been clinically approved for post-I/R neuroprotection. [...] Read more.
Brain ischemia-reperfusion (I/R) injury, commonly occurring in ischemic stroke and post-cardiac arrest scenarios, results in complex secondary damage involving oxidative stress, inflammation, apoptosis, and blood-brain barrier (BBB) breakdown. Despite decades of research, no pharmacological agent has yet been clinically approved for post-I/R neuroprotection. Natural compounds have recently gained attention for their multimodal therapeutic potential, including antioxidant, anti-inflammatory, anti-apoptotic, and neuroregenerative effects. This review highlights nine promising candidates—resveratrol, curcumin, quercetin, berberine, ginkgolide B, baicalin, naringin, fucoidan, and astaxanthin—that exhibit efficacy in experimental models of I/R injury when administered after the insult. Their chemical structures, pharmacokinetics, and mechanisms of action are described in detail, focusing on key signaling pathways such as nuclear factor erythroid 2-related (Nrf2), nuclear factor kappa B (NF-κB), phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt), and brain-derived neurotrophic factor (BDNF). Importantly, we outline the selection criteria for these compounds, including demonstrated neuroprotective efficacy, mechanistic clarity, and translational feasibility. While several challenges remain—such as limited bioavailability, BBB penetration, and species-specific metabolism—emerging strategies like nanoparticle delivery, synthetic analogs, and drug combinations offer potential solutions. By emphasizing the therapeutic versatility and mechanistic diversity of these natural agents, this review supports their clinical potential and encourages further preclinical optimization and biomarker-guided human trials. Full article
(This article belongs to the Section Neuroscience)
Show Figures

Graphical abstract

33 pages, 683 KB  
Review
Next-Gen Neuroprotection in Glaucoma: Synergistic Molecules for Targeted Therapy
by Alessio Martucci, Massimo Cesareo, Maria Dolores Pinazo-Durán, Francesco Aiello, Giulio Pocobelli, Raffaele Mancino and Carlo Nucci
J. Clin. Med. 2025, 14(17), 6145; https://doi.org/10.3390/jcm14176145 - 30 Aug 2025
Viewed by 334
Abstract
Background: Glaucoma is a progressive optic neuropathy marked by retinal ganglion cells (RGCs), apoptosis, vascular insufficiency, oxidative stress, mitochondrial dysfunction, excitotoxicity, and neuroinflammation. While intraocular pressure (IOP) reduction remains the primary intervention, many patients continue to lose vision despite adequate pressure control. Emerging [...] Read more.
Background: Glaucoma is a progressive optic neuropathy marked by retinal ganglion cells (RGCs), apoptosis, vascular insufficiency, oxidative stress, mitochondrial dysfunction, excitotoxicity, and neuroinflammation. While intraocular pressure (IOP) reduction remains the primary intervention, many patients continue to lose vision despite adequate pressure control. Emerging neuroprotective agents—citicoline, coenzyme Q10 (CoQ10), pyruvate, nicotinamide, pyrroloquinoline quinone (PQQ), homotaurine, berberine, and gamma-aminobutyric acid (GABA)—target complementary pathogenic pathways in experimental and clinical settings. Methods: This literature review synthesizes current evidence on glaucoma neuroprotection, specifically drawing on the most relevant and recent studies identified via PubMed. Results: Citicoline enhances phospholipid synthesis, stabilizes mitochondrial membranes, modulates neurotransmitters, and improves electrophysiological and visual field outcomes. CoQ10 preserves mitochondrial bioenergetics, scavenges reactive oxygen species, and mitigates glutamate-induced excitotoxicity. Pyruvate supports energy metabolism, scavenges reactive oxygen species, and restores metabolic transporter expression. Nicotinamide and its precursor nicotinamide riboside boost NAD+ levels, protect against early mitochondrial dysfunction, and enhance photopic negative response amplitudes. PQQ reduces systemic inflammation and enhances mitochondrial metabolites, while homotaurine modulates GABAergic signaling and inhibits β-amyloid aggregation. Berberine attenuates excitotoxicity, inflammation, and apoptosis via the P2X7 and GABA-PKC-α pathways. Preclinical models demonstrate synergy when agents are combined to address multiple targets. Clinical trials of fixed-dose combinations—such as citicoline + CoQ10 ± vitamin B3, citicoline + homotaurine ± vitamin E or PQQ, and nicotinamide + pyruvate—show additive improvements in RGCs’ electrophysiology, visual function, contrast sensitivity, and quality of life without altering IOP. Conclusions: A multi-targeted approach is suitable for glaucoma’s complex neurobiology and may slow progression more effectively than monotherapies. Ongoing randomized controlled trials are essential to establish optimal compound ratios, dosages, long-term safety, and structural outcomes. However, current evidence remains limited by small sample sizes, heterogeneous study designs, and a lack of long-term real-world data. Integrating combination neuroprotection into standard care holds promise for preserving vision and reducing the global burden of irreversible glaucoma-related blindness. Full article
(This article belongs to the Special Issue Advances in the Diagnosis and Treatment of Glaucoma)
Show Figures

Figure 1

31 pages, 4184 KB  
Article
Investigating the Neuroprotective, Hepatoprotective, and Antimicrobial Effects of Mushroom Extracts
by Menna-Allah E. Abdelkader, Hatungimana Mediatrice, Zhanxi Lin, Christopher Rensing, Mohamed M. Yacout, Dongmei Lin and Sarah A. Aggag
Int. J. Mol. Sci. 2025, 26(17), 8440; https://doi.org/10.3390/ijms26178440 - 29 Aug 2025
Viewed by 283
Abstract
Mushrooms, renowned for their nutritional value and bioactive compounds, offer potential health benefits, including antioxidants and anti-aging properties. Aging, characterized by cellular and tissue decline, is often associated with autophagy dysfunction, a crucial cellular cleaning process. This study aimed to investigate the neuroprotective, [...] Read more.
Mushrooms, renowned for their nutritional value and bioactive compounds, offer potential health benefits, including antioxidants and anti-aging properties. Aging, characterized by cellular and tissue decline, is often associated with autophagy dysfunction, a crucial cellular cleaning process. This study aimed to investigate the neuroprotective, hepatoprotective, and antimicrobial properties of extracts from four medicinal and edible mushrooms: Ganoderma lucidum, Hericium erinaceus, Pleurotus ostreatus, and Agaricus bisporus. The protein, total phenol, and flavonoid content of mushroom extracts were determined. Aging was induced with 120 mg/kg D-galactose and treated with 500 mg/kg mushroom extracts. The study evaluated liver enzyme levels, histopathological changes in liver and brain tissues, gene expression correlated to neurodegeneration (SEPT5-SV2B-ATXN2-PARK2), telomere length, and immunomodulatory and pro-inflammatory (IL-2-IL-4-IL-6) gene expression pathways. Additionally, the antimicrobial potential of mushroom extracts was assessed against several bacteria (Lysinibacillus odyssey, Lysinibacillus fusiformis, Klebsiella oxytoca, and Escherichia coli) using agar well diffusion and lowest minimum inhibitory concentration (MIC) methods. By exploring these diverse aspects, this study aimed to provide a foundation for a better understanding of the potential of mushrooms as natural neuroprotective, hepatoprotective, and antimicrobial agents and their potential applications in human health. Results indicated that all mushroom extracts effectively mitigated oxidative stress. Agaricus bisporus exhibited the highest protein and flavonoid content, and Pleurotus ostreatus displayed the highest phenolic content. Notably, Hericium erinaceus and Ganoderma lucidum extracts demonstrated significant neuroprotective and hepatoprotective properties against D-galactose-induced aging, as evidenced by histopathological examination. All extracts exhibited a significant decrease (p < 0.001) in liver function (serum levels of aspartate aminotransferase (GOT) and alanine aminotransferase (GPT)) and showed immunomodulatory and anti-inflammatory effects, characterized by upregulated IL-2 and IL-4 gene expression and downregulated IL-6 gene expression. Hericium erinaceus demonstrated the most pronounced upregulation (p < 0.001) of SEPT5, SV2B, and telomere length gene expression, suggesting potential anti-aging effects. Furthermore, all mushroom extracts displayed antimicrobial activity against the tested bacterial strains, except Hericium erinaceus, which exhibited antibacterial activity solely against E. coli. Agaricus bisporus exhibited the largest inhibition zones (22 ± 0.06 mm) against Lysinibacillus odyssey, while Hericium erinaceus displayed the largest inhibition zone against E. coli. The MIC value was observed with Agaricus bisporus extract against Lysinibacillus odyssey (1.95 ± 0.16 mg/mL). Lysinibacillus fusiformis exhibited the highest resistance to the tested mushroom extracts. These findings suggest that these edible and medicinal mushrooms possess a wide range of health-promoting properties, including neuroprotective, hepatoprotective, and antimicrobial activities. Further research is needed to fully understand the underlying mechanisms and optimize applications. However, our results provide a strong foundation for exploring these mushrooms as potential natural agents that promote overall health and combat age-related decline. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
Show Figures

Figure 1

25 pages, 662 KB  
Review
Anti-Inflammatory and Neuroprotective Effects of Undaria pinnatifida Fucoidan
by Cheng Yang, Corinna Dwan, Barbara C. Wimmer, Sayed Koushik Ahamed, Fionnghuala James, Jigme Thinley, Richard Wilson, Luke Johnson and Vanni Caruso
Mar. Drugs 2025, 23(9), 350; https://doi.org/10.3390/md23090350 - 29 Aug 2025
Viewed by 539
Abstract
Undaria pinnatifida fucoidan (UPF), a sulphated polysaccharide derived from brown seaweed, has attracted scientific and clinical interest for its wide-ranging anti-inflammatory and neurodegenerative properties. A growing body of research shows that UPF inhibits NF-κB and MAPK signalling pathways, reduces pro-inflammatory cytokines (TNF-α, IL-1β, [...] Read more.
Undaria pinnatifida fucoidan (UPF), a sulphated polysaccharide derived from brown seaweed, has attracted scientific and clinical interest for its wide-ranging anti-inflammatory and neurodegenerative properties. A growing body of research shows that UPF inhibits NF-κB and MAPK signalling pathways, reduces pro-inflammatory cytokines (TNF-α, IL-1β, IL-6), decreases ROS production, and suppresses iNOS and COX-2 activity, thereby mitigating oxidative and inflammatory damage in vitro. In vivo studies confirm these actions, demonstrating reduced systemic inflammation, promoted antioxidant defence, modulated gut microbiota composition, and improved production of beneficial microbial metabolites. In parallel, emerging evidence highlights UPF’s neuroprotective potential, characterised by protection against neuroinflammation and oxidative stress, the attenuation of amyloid-beta deposition, and improvement in neuronal function. Importantly, low- to medium-molecular-weight and highly sulphated UPF fractions consistently exhibit stronger bioactivities, suggesting a structural basis for its therapeutic potential. This review integrates mechanistic evidence from cellular, preclinical, and emerging clinical studies, highlighting UPF as a versatile marine-derived agent with therapeutic relevance for inflammatory and neurodegenerative diseases, and outlines future research directions toward clinical translation. Full article
Show Figures

Figure 1

19 pages, 1366 KB  
Review
Ketamine’s Role in Neuroinflammation and Neuroprotection Across Neurological and Psychiatric Disorders: A Narrative Review
by Gustavo N. Silva, Virna G. A. Brandão, Kenneth Blum, Kai-Uwe Lewandrowski and Rossano K. A. Fiorelli
Pharmaceuticals 2025, 18(9), 1298; https://doi.org/10.3390/ph18091298 - 29 Aug 2025
Viewed by 316
Abstract
Ketamine, a widely used anesthetic with emerging evidence suggesting neuroprotective and anti-inflammatory properties across various neurological disorders, is recognized for its NMDA receptor antagonism. It has been postulated to play a role in neuroprotection, due to its anti-inflammatory properties, and decrease microglial activation, [...] Read more.
Ketamine, a widely used anesthetic with emerging evidence suggesting neuroprotective and anti-inflammatory properties across various neurological disorders, is recognized for its NMDA receptor antagonism. It has been postulated to play a role in neuroprotection, due to its anti-inflammatory properties, and decrease microglial activation, as well as cytokines TNF and IL-6. Despite its established role, the extent of ketamine’s effects on neuroinflammation and neuroprotection remains to be fully elucidated. Here, we conducted a narrative review synthesizing current knowledge on ketamine’s operating mechanisms, including its modulation of synaptic plasticity, excitotoxicity, and cytokine release, alongside its therapeutic applications in traumatic brain injury, neurodegenerative diseases, psychiatric disorders, and pain management. For this narrative review, we searched the Medline, Embase, Scopus, Web of Science, and PubMed databases. Our findings indicate that ketamine reduces excitotoxicity and inflammation, which may contribute to neuroprotection in acute neurological injuries. These insights underscore ketamine’s potential as an adjunctive neuroprotective agent, warranting further clinical investigation to optimize its therapeutic utility across neurological and psychiatric contexts. Full article
(This article belongs to the Special Issue Pharmacological Insight into NMDA Receptor Antagonists)
Show Figures

Figure 1

25 pages, 2736 KB  
Article
Therapeutic Effects of Intranasal Administration of Mesenchymal Stem Cell-Derived Secretome in Rats Exposed to Chronic Unpredictable Mild Stress
by Alba Ávila, María Eugenia Riveros, Sofía Adasme, Coram Guevara, Rodrigo Del Rio, Fernando C. Ortiz, Nicole Leibold and Fernando Ezquer
Pharmaceutics 2025, 17(9), 1129; https://doi.org/10.3390/pharmaceutics17091129 - 29 Aug 2025
Viewed by 427
Abstract
Background: Major depression is a significant source of suffering and economic loss. Despite efforts to understand this condition and find better treatments, the burden imposed by this disease continues to rise. Most approved pharmacological treatments for depression focus on controlling the availability [...] Read more.
Background: Major depression is a significant source of suffering and economic loss. Despite efforts to understand this condition and find better treatments, the burden imposed by this disease continues to rise. Most approved pharmacological treatments for depression focus on controlling the availability of monoamines in synapses. However, accumulating evidence suggests that neuroinflammation, oxidative stress, and reduced hippocampal neurogenesis play key roles as causal factors in the development of major depression symptoms. Therefore, preclinical testing of pharmacological approaches targeting these factors is essential. Mesenchymal stem cells (MSCs) are known for their potential as powerful antioxidants and anti-inflammatory agents, exerting neuroprotective actions in the brain. They produce various therapeutic molecules in a paracrine manner, collectively known as secretome. Methods: In this work, we evaluated the antidepressant potential of repeated intranasal administration of MSC-derived secretome in an animal model of major depressive disorder induced by chronic mild unpredictable stress. Results: We observed that intranasal administration of MSC-derived secretome reduced the appearance of some of the behavioral parameters commonly associated with major depression, including anhedonic, apathetic, and anxious behaviors, inducing a strong reduction in the overall depression score compared to vehicle-treated animals. At the structural level, secretome administration prevented increased astrocyte density and the atrophy of astrocyte processes observed in vehicle-treated stressed animals. Additionally, secretome administration induced an increase in myelin levels and oligodendroglia in the cortex. Conclusions: Our data suggests that intranasal administration of MSC-derived secretome may represent a potential therapeutic alternative to current treatments for this devastating pathology. Full article
Show Figures

Graphical abstract

26 pages, 1102 KB  
Review
HDACs in the Brain: From Chromatin Remodeling to Neurodegenerative Disease
by Luan Pereira Diniz, Pedro de Sena Murteira Pinheiro, Lucas S. Franco and Flávia Carvalho Alcantara Gomes
Cells 2025, 14(17), 1338; https://doi.org/10.3390/cells14171338 - 29 Aug 2025
Viewed by 474
Abstract
Histone deacetylases (HDACs) are key epigenetic regulators that influence chromatin remodeling, gene expression, and cellular plasticity in the central nervous system (CNS). This review provides a comprehensive overview of the classification and functional diversity of HDACs, with particular emphasis on their roles in [...] Read more.
Histone deacetylases (HDACs) are key epigenetic regulators that influence chromatin remodeling, gene expression, and cellular plasticity in the central nervous system (CNS). This review provides a comprehensive overview of the classification and functional diversity of HDACs, with particular emphasis on their roles in neural progenitor cells, mature neurons, and glial populations. In neural stem and progenitor cells, HDACs modulate neurogenesis, fate specification, and lineage commitment. In differentiated neurons, HDACs govern synaptic plasticity, memory formation, and survival. In glial cells, including astrocytes and microglia, HDACs orchestrate inflammatory responses, redox balance, and metabolic adaptations. We further examine the dysregulation of HDAC expression and activity in major neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease. Evidence from human post-mortem brain studies reveals region- and isoform-specific alterations in HDAC expression, which are closely associated with cognitive decline, mitochondrial dysfunction, and neuroinflammation. Preclinical studies support the use of HDAC inhibitors (HDACi) as neuroprotective agents, capable of restoring acetylation homeostasis, reducing neuroinflammation, and improving neuronal function. Given the relevance of HDACi, we summarize current clinical studies assessing the safety of these compounds in the context of tumor biology, as well as their potential future applications in neurodegenerative diseases. Together, this review underscores the dual significance of HDACs as biomarkers and therapeutic targets in the context of CNS disorders. Full article
Show Figures

Figure 1

14 pages, 268 KB  
Review
Beyond the Eye: Glaucoma and the Brain
by Marco Zeppieri, Federico Visalli, Mutali Musa, Alessandro Avitabile, Rosa Giglio, Daniele Tognetto, Caterina Gagliano, Fabiana D’Esposito and Francesco Cappellani
Brain Sci. 2025, 15(9), 934; https://doi.org/10.3390/brainsci15090934 - 28 Aug 2025
Viewed by 419
Abstract
Glaucoma is traditionally classified as an ocular disease characterized by progressive retinal ganglion cell (RGC) loss and optic nerve damage. However, emerging evidence suggests that its pathophysiology may extend beyond the eye, involving trans-synaptic neurodegeneration along the visual pathway and structural changes within [...] Read more.
Glaucoma is traditionally classified as an ocular disease characterized by progressive retinal ganglion cell (RGC) loss and optic nerve damage. However, emerging evidence suggests that its pathophysiology may extend beyond the eye, involving trans-synaptic neurodegeneration along the visual pathway and structural changes within central brain regions, including the lateral geniculate nucleus and visual cortex. In this narrative review, we have used the phrase ‘brain involvement’ to underscore central changes that accompany or follow retinal ganglion cell loss; we have not intended to redefine glaucoma as a primary cerebral disorder. Neuroimaging studies and neurocognitive assessments in adult glaucoma patients, primarily older individuals with primary open-angle glaucoma reveal that glaucoma patients may exhibit alterations in brain connectivity and cortical thinning, aligning it more closely with neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease. This evolving neurocentric perspective raises important questions regarding shared mechanisms—such as mitochondrial dysfunction, chronic inflammation, and impaired axonal transport—that may link glaucomatous optic neuropathy to central nervous system (CNS) pathology. These insights open promising therapeutic avenues, including the repurposing of neuroprotective and neuroregenerative agents, targeting not only intraocular pressure (IOP) but also broader CNS pathways. Furthermore, neuroimaging biomarkers and brain-targeted interventions may play a future role in diagnosis, prognosis, and individualized treatment. This review synthesizes current evidence supporting glaucoma as a CNS disease, explores the mechanistic overlap with neurodegeneration, and discusses the potential clinical implications of glaucoma within a neuro-ophthalmologic paradigm. Full article
23 pages, 4058 KB  
Article
Inhibition of Astrocyte Reactivity by Mdivi-1 After Status Epilepticus in Rats Exacerbates Microglia-Mediated Neuroinflammation and Impairs Limbic–Cortical Glucose Metabolism
by Francisca Gómez-Oliver, Rubén Fernández de la Rosa, Mirjam Brackhan, Pablo Bascuñana, Miguel Ángel Pozo and Luis García-García
Biomolecules 2025, 15(9), 1242; https://doi.org/10.3390/biom15091242 - 27 Aug 2025
Viewed by 350
Abstract
The lithium–pilocarpine rat model of status epilepticus (SE) is a well-established paradigm for studying epileptogenesis. Astrocyte reactivity has been implicated in modulating seizure susceptibility and neuroinflammation, yet its functional role in early epileptogenesis remains unclear. Herein, we evaluated the effects of Mdivi-1, a [...] Read more.
The lithium–pilocarpine rat model of status epilepticus (SE) is a well-established paradigm for studying epileptogenesis. Astrocyte reactivity has been implicated in modulating seizure susceptibility and neuroinflammation, yet its functional role in early epileptogenesis remains unclear. Herein, we evaluated the effects of Mdivi-1, a pharmacological inhibitor of mitochondrial fission protein Drp1, for its ability to modulate astrocytic mitochondrial dynamics and for its reported preventive neuroprotective properties. Mdivi-1 was administered shortly after SE onset, and we assessed brain glucose metabolism using [18F]FDG PET, alongside histological markers of neurodegeneration, astrocyte reactivity, and microglial activation, at 3 days post-SE. As expected, SE induced widespread brain hypometabolism measured by a VOI analysis, hippocampal neurodegeneration, and glial activation. Post-SE Mdivi-1 administration reduced hippocampal astrogliosis but neither conferred neuroprotection nor rescued glucose metabolism. On the contrary, Mdivi-1 exacerbated limbic–cortical hypometabolism when evaluated by SPM and normalized to whole brain tracer uptake and microglia-mediated neuroinflammation. These findings challenge the assumption that early astrocyte inhibition confers neuroprotection. Furthermore, early suppression of astrocyte reactivity after the damage has occurred may shift the neuroinflammatory response toward maladaptive microglial activation. Thus, while Mdivi-1 holds promise as a preventive neuroprotective agent, its use post-SE may have unintended adverse effects on the brain’s response to SE. Full article
(This article belongs to the Special Issue Biomolecular Approaches and Drugs for Neurodegeneration—2nd Edition)
Show Figures

Figure 1

Back to TopTop