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
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
remove_circle_outline

Search Results (1,881)

Search Parameters:
Keywords = brain toxicity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 2754 KB  
Article
Propofol-Induced Mitochondrial Dysfunction Is Independent of Mitochondrial Permeability Transition
by Aya Kawachi, Shoichiro Shibata, Eskil Elmér and Hiroyuki Uchino
Biomedicines 2025, 13(12), 3125; https://doi.org/10.3390/biomedicines13123125 (registering DOI) - 18 Dec 2025
Abstract
Background/Objectives: In recent years, it has been suggested that sedatives may cause brain damage. One possible mechanism is interference with oxidative phosphorylation of brain mitochondria, but much remains unknown. In this study, we focused on dexmedetomidine, midazolam, and propofol, essential sedatives in [...] Read more.
Background/Objectives: In recent years, it has been suggested that sedatives may cause brain damage. One possible mechanism is interference with oxidative phosphorylation of brain mitochondria, but much remains unknown. In this study, we focused on dexmedetomidine, midazolam, and propofol, essential sedatives in anesthesia and intensive care, and aimed to understand the effects of these drugs on mouse brain mitochondria. Methods: We measured changes in mitochondrial respiratory capacity and swelling rate upon exposure to these sedatives in a wide concentration range. For the sedative that demonstrated impaired mitochondrial function we explored the possible involvement of mitochondrial permeability transition pore opening using brain mitochondria from cyclophilin D knockout (CypD KO) mice and detected cytochrome c (cyt c) release by Western blot. Results: Of the three sedatives, only high concentrations of propofol exhibited reduced respiratory capacity and mitochondrial swelling, toxicity which was not prevented by CypD KO. Furthermore, propofol did not induce cyt c release. Conclusions: These results suggest that propofol-induced brain mitochondrial dysfunction is a mechanism independent of mPTP opening. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Graphical abstract

20 pages, 625 KB  
Review
Pathways, Mechanisms, and Therapeutic Strategies of Neurotoxicity Induced by Micro- and Nanoplastics
by Min Yan, Yanfeng Chen, Ying Tao, Hui Wang, Xuewen Tian and Xiuxiu Wang
Brain Sci. 2025, 15(12), 1345; https://doi.org/10.3390/brainsci15121345 - 18 Dec 2025
Abstract
Plastic pollution now represents a global environmental crisis, as micro- and nanoplastics (MNPs) infiltrate organisms via multiple routes such as the digestive system and respiratory system, ultimately accumulating in tissues and endangering human health. The central nervous system exhibits particular vulnerability to MNPs [...] Read more.
Plastic pollution now represents a global environmental crisis, as micro- and nanoplastics (MNPs) infiltrate organisms via multiple routes such as the digestive system and respiratory system, ultimately accumulating in tissues and endangering human health. The central nervous system exhibits particular vulnerability to MNPs toxicity, which can trigger neurotoxic effects and disrupt brain function, potentially contributing to neurological disorders. Understanding the precise mechanisms and biological pathways underlying MNP-induced neurotoxicity has therefore emerged as a critical step toward identifying therapeutic targets. This review synthesizes current knowledge on MNPs’ entry routes into the brain, examines proposed neurotoxic mechanisms, and evaluates existing and prospective treatment approaches. These insights may inform both the risk assessment of MNPs exposure and the development of targeted interventions for neurological protection. Full article
(This article belongs to the Topic Healthy, Safe and Active Aging, 2nd Edition)
Show Figures

Figure 1

18 pages, 313 KB  
Review
Underlying Mechanisms of GBA1 in Parkinson’s Disease and Dementia with Lewy Bodies: Narrative Review
by Anastasia Bougea
Genes 2025, 16(12), 1496; https://doi.org/10.3390/genes16121496 - 15 Dec 2025
Viewed by 183
Abstract
Background/Objectives: Parkinson’s disease (PD) and Dementia with Lewy Bodies (DLB) are neurodegenerative disorders characterized by the accumulation of misfolded alpha-synuclein protein in the brain. Mutations in the glucocerebrosidase 1 (GBA1) gene have been identified as a significant genetic risk factor [...] Read more.
Background/Objectives: Parkinson’s disease (PD) and Dementia with Lewy Bodies (DLB) are neurodegenerative disorders characterized by the accumulation of misfolded alpha-synuclein protein in the brain. Mutations in the glucocerebrosidase 1 (GBA1) gene have been identified as a significant genetic risk factor for both PD and DLB. GBA1 encodes for the lysosomal enzyme glucocerebrosidase, which is responsible for the breakdown of glucosylceramide (GC). Deficiencies in glucocerebrosidase activity lead to the accumulation of glucosylceramide within lysosomes, contributing to lysosomal dysfunction and impaired protein degradation. The aim of this narrative review is to update the underlying mechanisms by which GBA1 mutations contribute to the pathogenesis of PD and DLB. Methods: A comprehensive literature search was conducted across four major electronic databases (PubMed, Web of Science (Core Collection), Scopus, and Embase) from inception to 8 November 2025. The initial search identified approximately 1650 articles in total, with the number of hits from each database being as follows: PubMed (~450), Web of Science (~380), Scopus (~520), and Embase (~300). Results: The mechanism by which mutations in the GBA1 gene contribute to PD involves both loss-of- function and gain-of-function pathways, which are not mutually exclusive. Typically, GBA1 mutations lead to a loss of function by reducing the activity of the GCase enzyme, impairing the autophagy- lysosomal pathway and leading to α-synuclein accumulation. However, some mutant forms (GBA1L444P) of the GCase enzyme can also acquire a toxic gain of function, contributing to α-synuclein aggregation through mechanisms like endoplasmic reticulum stress and misfolding. While Venglustat effectively reduced GC levels, a key marker associated with GBA1-PD, the lack of clinical improvement led to the discontinuation of its development for this indication. Conclusions: GBA1-mediated lysosomal and lipid dysregulation represents a key pathogenic axis in PD and DLB. Understanding these mechanisms provides crucial insight into disease progression and highlights emerging therapeutic strategies—such as pharmacological chaperones, substrate reduction therapies, and gene-targeted approaches—aimed at restoring GCase function and lysosomal homeostasis to slow or prevent neurodegeneration. Full article
(This article belongs to the Special Issue Genetics and Epigenetics in Neurological Disorders)
20 pages, 1160 KB  
Review
Prenatal Exposure to Tobacco Smoke and Vaping Aerosols: Mechanisms Disrupting White-Matter Formation
by Sebastián Beltran-Castillo, Juan Pablo Espinoza and Michelle Grambs
Toxics 2025, 13(12), 1071; https://doi.org/10.3390/toxics13121071 - 12 Dec 2025
Viewed by 190
Abstract
White-matter development during fetal life represents one of the most vulnerable processes to environmental disruption, as it relies on the precisely timed proliferation, migration, and differentiation of oligodendrocyte lineage cells. Among environmental threats, exposure to toxic compounds contained in tobacco smoke and vaping [...] Read more.
White-matter development during fetal life represents one of the most vulnerable processes to environmental disruption, as it relies on the precisely timed proliferation, migration, and differentiation of oligodendrocyte lineage cells. Among environmental threats, exposure to toxic compounds contained in tobacco smoke and vaping aerosols represents a major yet preventable risk during pregnancy. Despite growing awareness, tobacco smoking remains widespread, and a substantial proportion of the population—including pregnant women—continues to perceive electronic nicotine delivery systems (ENDS) as less harmful, a misconception that contributes to persistent prenatal exposure. These products expose the fetus to numerous substances that readily cross the placenta and reach the developing brain, including compounds with endocrine-disrupting activity, where they interfere with white-matter development. Epidemiological and neuroimaging studies consistently reveal microstructural alterations in white matter that correlate with long-term cognitive and behavioral impairments in offspring exposed in utero. These alterations may arise from both nicotine-specific pathways and the actions of other toxicants in cigarette smoke and ENDS aerosols that cross the placenta and disrupt white-matter emergence and maturation. Preclinical research provides mechanistic insight: nicotine acts directly on nicotinic acetylcholine receptors (nAChRs) in oligodendrocyte precursor cells, disrupting calcium signaling and differentiation, while additional constituents of smoke and vaping aerosols also affect astrocyte and microglial function and disturb the extracellular milieu required for proper myelination. Full article
(This article belongs to the Special Issue Reproductive and Developmental Toxicity of Environmental Factors)
Show Figures

Graphical abstract

21 pages, 3412 KB  
Article
Generation and Characterization of Novel Contilisant+Tubastatin a Multitarget Small Molecules Against Glioblastoma
by Irati de Goñi, Aizpea Artetxe-Zurutuza, Joseba Elizazu, Raul Garcia-Garcia de Garayo, Jhonatan Vergara-Arce, Mikel Azkargorta, Mireia Toledano-Pinedo, Alicia Porro-Pérez, Felix Elortza, Jose Luis Marco-Contelles, Nicolas Sampron, Nerea Iturrioz-Rodriguez and Ander Matheu
Pharmaceutics 2025, 17(12), 1594; https://doi.org/10.3390/pharmaceutics17121594 - 10 Dec 2025
Viewed by 273
Abstract
Background/Objectives: Glioblastoma is the most common and aggressive primary brain tumor in adults, with patient prognosis remaining poor. Treatment resistance and tumor recurrence are frequent, primarily due to the high intra- and inter-tumoral heterogeneity and the existence of glioma stem cells. Thus, [...] Read more.
Background/Objectives: Glioblastoma is the most common and aggressive primary brain tumor in adults, with patient prognosis remaining poor. Treatment resistance and tumor recurrence are frequent, primarily due to the high intra- and inter-tumoral heterogeneity and the existence of glioma stem cells. Thus, there is an urgent need for novel and more effective therapeutic strategies. Multitarget small molecules (MSMs) are emerging as a novel therapeutic strategy for the treatment of complex diseases such as cancer. Methods: In the present work, we have generated a novel family of indole-based MSMs with pharmacophoric moieties combining the parent compounds Contilisant and the HDAC inhibitor Tubastatin A. Thus, the MSMs were designed to inhibit monoamine oxidases (MAOs), cholinesterases (ChEs) and histone deacetylases (HDACs), while acting as histamine H3 receptor (H3R) antagonists and sigma 1 receptor (S1R) agonists. We generated four different molecules and evaluated in detail the activity of the two most efficient MSM compounds in vitro and in vivo. Results: These molecules induced potent cytotoxic effects in vitro in patient-derived glioma stem cells and glioblastoma cell lines and significantly impaired tumor growth in vivo. OMIC analyses further revealed that the compounds induce dysregulation of the cell cycle in glioma stem cells. Moreover, in silico analyses indicated that these compounds are theoretically capable of crossing the blood–brain barrier, while exhibiting low toxicity in healthy cells. Conclusions: In conclusion, our findings demonstrate the potential antitumor activity of a novel family of MSMs in preclinical models of glioblastoma. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Figure 1

33 pages, 6086 KB  
Article
Disturbances in Mitochondrial Network, Biogenesis, and Mitochondria-Mediated Inflammatory Responses in Selected Brain Structures of Rats Exposed to Lead (Pb) During Prenatal and Neonatal Development
by Mikołaj Chlubek, Magdalena Gąssowska-Dobrowolska, Agnieszka Kolasa, Maciej Tarnowski, Patrycja Tomasiak, Agnieszka Maruszewska, Katarzyna Barczak and Irena Baranowska-Bosiacka
Int. J. Mol. Sci. 2025, 26(24), 11907; https://doi.org/10.3390/ijms262411907 - 10 Dec 2025
Viewed by 149
Abstract
Lead (Pb) disrupts mitochondrial function, but its impact on the mitochondrial dynamics and biogenesis during early brain development remains insufficiently understood. This study aimed to investigate the effects of pre- and neonatal Pb exposure on the processes involved in mitochondrial network formation in [...] Read more.
Lead (Pb) disrupts mitochondrial function, but its impact on the mitochondrial dynamics and biogenesis during early brain development remains insufficiently understood. This study aimed to investigate the effects of pre- and neonatal Pb exposure on the processes involved in mitochondrial network formation in the brains of rat offspring, simulating environmental exposure. We quantified mRNA expression (qRT-PCR) and protein levels (ELISA) of key mitochondrial fusion (Mfn1, Mfn2, Opa1), fission (Drp1, Fis1) regulators, as well as biogenesis markers (PGC-1α, TFAM, NRF1) in the hippocampus, forebrain cortex, and cerebellum of rats exposed to Pb. Mitochondrial ultrastructure was evaluated using transmission electron microscopy (TEM), and the expression of mitochondrial electron transport chain (ETC) genes was analysed (qRT-PCR). Furthermore, to examine the involvement of the cGAS–STING pathway in Pb-induced neuroinflammation, we measured the expression of ISGs (qRT-PCR), TBK1 phosphorylation (Western blot), and 2′,3′-cGAMP synthesis (ELISA). Our results showed that Pb exposure markedly reduced PGC-1α and region-specific NRF1 levels, broadly supressed fusion proteins (Mfn1, Mfn2, Opa1), increased Fis1, and depleted Drp1. ETC gene expression (mtNd1, mtCyb and mtCo1) were upregulated in a brain-structure-dependent manner. These molecular changes were accompanied by pronounced mitochondrial morphological abnormalities. Despite upregulation of Mx1, Ifi44, and Sting1, along with synthesis of 2′3′-cGAMP, TBK1 activation was not detected. All these findings demonstrate that early-life Pb exposure, even low-dose, disrupts mitochondrial biogenesis and the fusion–fission machinery, thus impairs brain energy homeostasis, and implicates mitochondria as central mediators of Pb-induced neuroinflammation and neurodevelopmental toxicity. Full article
(This article belongs to the Special Issue Unraveling the Molecular Mechanisms of Neurodegeneration)
Show Figures

Figure 1

22 pages, 5118 KB  
Article
Investigating Nickel-Induced Neurotoxicity: Associations with Gut Microbiota Dysbiosis and Ferroptosis
by Yao Shen, Kai Cao, Wenjuan Zhang, Chun Chen, Chang Gao, Jingran Wang, Tian Xin, Cun Li, Shusheng Tang, Xingyao Pei and Daowen Li
Antioxidants 2025, 14(12), 1478; https://doi.org/10.3390/antiox14121478 - 9 Dec 2025
Viewed by 333
Abstract
Nickel is a pervasive heavy metal with the potential for multi-route exposure, raising significant concerns regarding systemic toxicity. Although Ni2+ has been implicated in nickel sulfate NiSO4-induced neurotoxicity, its underlying mechanisms remain incompletely elucidated. The present study investigates the role [...] Read more.
Nickel is a pervasive heavy metal with the potential for multi-route exposure, raising significant concerns regarding systemic toxicity. Although Ni2+ has been implicated in nickel sulfate NiSO4-induced neurotoxicity, its underlying mechanisms remain incompletely elucidated. The present study investigates the role of NiSO4-induced ferroptosis as a potential contributor to neurotoxicity. C57BL/6 mice were administered NiSO4 daily via oral gavage at doses of 50, 100, and 200 mg/kg over 28 days. Neurobehavioral assessments, histopathological examination, transmission electron microscopy, and molecular profiling were conducted to evaluate brain injury and ferroptotic activity. Gut microbiota composition and intestinal barrier integrity were systematically evaluated. In vitro, HT22 cells were subjected to NiSO4 treatment, followed by integrative transcriptomic analysis complemented by pharmacological and genetic manipulation to delineate the contributions of ferroptosis and autophagy. The results demonstrated that NiSO4 exposure inhibited body weight gain, elicited depression-like behaviors, and initiated ferroptosis, evidenced by ultrastructural mitochondrial damage and dysregulated expression of glutathione peroxidase 4/acyl-CoA synthetase long chain family member 4 (GPX4/ACSL4). Furthermore, NiSO4 caused gut microbiota dysbiosis and compromised the intestinal barrier, which was correlated with the induction of ferroptosis in neuronal cells of the brain. In HT22 cells, NiSO4 elicited dose-dependent cytotoxicity and lactate dehydrogenase (LDH) release. KEGG pathway enrichment analysis further revealed that NiSO4 treatment significantly upregulated pathways associated with ferroptosis, autophagy, and lysosomal function. Moreover, both ferrostatin-1 and rapamycin attenuated NiSO4-induced cytotoxicity and ferroptosis, indicating that autophagy serves a protective function against ferroptotic cell death. Additionally, overexpression of Transcription Factor EB (TFEB) attenuated NiSO4-induced ferroptosis by downregulating ACSL4, and upregulating GPX4, implicating the autophagy–lysosome pathway in the protective regulation of this cell death process. In summary, our findings indicated that NiSO4-induced neurotoxicity was strongly associated with gut microbiota dysbiosis and coincided with ferroptosis in the brain, while stimulation of the autophagy–lysosome pathway conferred neuroprotective effects via modulating TFEB-dependent anti-ferroptotic mechanisms. These findings offer novel insights for risk assessment and therapeutic strategies of nickel-related neurotoxicity. Full article
Show Figures

Figure 1

33 pages, 2706 KB  
Review
Targeting Cathepsins in Neurodegeneration: Biochemical Advances
by Francesca Di Matteo, Mariapia Vietri, Simone D’Alessio, Tania Ciaglia, Erica Federica Vestuto, Giacomo Pepe, Ornella Moltedo, Veronica Di Sarno, Simona Musella, Carmine Ostacolo, Fabio Cominelli, Pietro Campiglia, Alessia Bertamino, Maria Rosaria Miranda and Vincenzo Vestuto
Biomedicines 2025, 13(12), 3019; https://doi.org/10.3390/biomedicines13123019 - 9 Dec 2025
Viewed by 228
Abstract
Background/Objectives: Cathepsins, lysosomal proteases crucial for neuronal proteostasis, mediate the clearance of misfolded and aggregated proteins. Their dysregulation is implicated in neurodegenerative and neuropsychiatric disorders such as Alzheimer’s, Parkinson’s, and Huntington’s diseases. These conditions are characterized by toxic protein accumulation and impaired [...] Read more.
Background/Objectives: Cathepsins, lysosomal proteases crucial for neuronal proteostasis, mediate the clearance of misfolded and aggregated proteins. Their dysregulation is implicated in neurodegenerative and neuropsychiatric disorders such as Alzheimer’s, Parkinson’s, and Huntington’s diseases. These conditions are characterized by toxic protein accumulation and impaired clearance, which exacerbate cellular stress responses, including the unfolded protein response (UPR), oxidative damage, and mitochondrial dysfunction. This review aims to summarize current knowledge on cathepsin roles in these pathways and assess their therapeutic potential. Methods: A comprehensive literature review was conducted, focusing on recent in vitro and in vivo studies investigating cathepsin function, inhibition, and modulation. Mechanistic insights and pharmacological approaches targeting cathepsins were analyzed, with attention to challenges in translating preclinical findings to clinical settings. Results: Cathepsins demonstrate a dual role: their proteolytic activity supports neuronal health by degrading toxic aggregates, but altered or insufficient activity may worsen proteotoxic stress. Studies reveal that cathepsins regulate autophagy, apoptosis, and neuroinflammation both intracellularly and extracellularly. Despite promising mechanistic data, clinical translation is hindered by issues such as poor inhibitor selectivity, limited brain penetration, and variability across preclinical models. Conclusions: Targeting cathepsins presents a promising strategy for treating neurodegenerative and neuropsychiatric disorders, but significant challenges remain. Future research should focus on improving drug specificity and delivery, and on developing standardized models to better predict clinical outcomes. Full article
Show Figures

Figure 1

19 pages, 674 KB  
Review
Natural Anti-Inflammatory Agents in the Chemoprevention of Gliomas: Targeting Neuroinflammation and the Tumor Microenvironment
by Georgios S. Markopoulos, Chrissa Sioka, George A. Alexiou, Dimitrios Peschos, George Vartholomatos and Athanassios P. Kyritsis
Cancers 2025, 17(24), 3922; https://doi.org/10.3390/cancers17243922 - 8 Dec 2025
Viewed by 293
Abstract
Chronic inflammation is a well-established hallmark of cancer, playing a critical role in the initiation and progression of gliomas. Recent evidence underscores the importance of anti-inflammatory natural products as chemotherapeutic and potentially as chemopreventive agents, offering a safe and multifaceted approach to mitigate [...] Read more.
Chronic inflammation is a well-established hallmark of cancer, playing a critical role in the initiation and progression of gliomas. Recent evidence underscores the importance of anti-inflammatory natural products as chemotherapeutic and potentially as chemopreventive agents, offering a safe and multifaceted approach to mitigate tumor-promoting inflammation in the brain. This review explores the interplay between major inflammation-related pathways—such as NF-κB, COX-2, and the NLRP3 inflammasome—and key bioactive compounds derived from natural sources such as polyphenols, isothiocyanates, terpenes/lignans, and omega-3-derived mediators. We provide evidence on the effect of these compounds on the above inflammatory triangle. We discuss emerging in vitro, in vivo preclinical and translational evidence in the context of glioma biology and highlight how these compounds may pass the blood–brain barrier (BBB) and modulate the tumor microenvironment (TME), including immune cell infiltration and cytokine profiles that may act in a pro- or anti-inflammatory manner, highlighting their capacity to inhibit GBM-associated inflammation. Each substance may differentially influence the components of the inflammatory triangle. Overall, we position these agents as low-toxicity, formulation-aware adjuncts to standard care. The ultimate goal is offering novel insights on low-toxicity, inflammation-targeting interventions against malignant brain tumors. Full article
(This article belongs to the Special Issue Chemoprevention Advances in Cancer (2nd Edition))
Show Figures

Figure 1

25 pages, 1362 KB  
Review
Emerging Frontiers in Neuro-Oncology: Insights into Extracellular Vesicle-Driven Tumor Mechanisms and Nanotherapeutic Strategies
by Tommaso Colangelo, Anna Alessia Saponaro, Gianluigi Mazzoccoli, Gaetano Serviddio and Rosanna Villani
Int. J. Mol. Sci. 2025, 26(24), 11826; https://doi.org/10.3390/ijms262411826 - 7 Dec 2025
Viewed by 198
Abstract
Brain tumors encompass a heterogeneous group of neoplasms, including primary and secondary metastatic lesions, with glioblastoma multiforme (GBM) representing the most aggressive primary malignancy. Despite advancements in surgical resection, radiotherapy, and chemotherapy, the prognosis for GBM remains poor due to its infiltrative nature, [...] Read more.
Brain tumors encompass a heterogeneous group of neoplasms, including primary and secondary metastatic lesions, with glioblastoma multiforme (GBM) representing the most aggressive primary malignancy. Despite advancements in surgical resection, radiotherapy, and chemotherapy, the prognosis for GBM remains poor due to its infiltrative nature, tumor heterogeneity and resistance mechanisms. Emerging diagnostic tools, such as liquid biopsies, and therapeutic strategies leveraging extracellular vesicles (EVs) are reshaping the field of neuro-oncology. EVs, lipid bilayer-enclosed particles secreted by cells, carry oncogenic cargo such as microRNAs and molecular chaperones, influencing tumor progression, immune evasion, and therapy resistance. Recent research highlights their potential as biomarkers for early diagnosis and vehicles for targeted drug delivery across the blood–brain barrier (BBB). EV-based nanotherapeutics show promise in improving treatment precision, reducing systemic toxicity, and advancing precision medicine in brain tumor management. However, challenges related to EV heterogeneity, cargo-loading efficiency, and large-scale production must be addressed to fully realize their therapeutic potential. This review explores the multifaceted roles of EVs in brain tumors, emphasizing their diagnostic, prognostic, and therapeutic applications. Full article
Show Figures

Figure 1

31 pages, 1933 KB  
Review
The Applications of Nanocellulose and Its Modulation of Gut Microbiota in Relation to Obesity and Diabetes
by Tai L. Guo, Ayushi Bhagat and Daniel J. Guo
J. Nanotheranostics 2025, 6(4), 34; https://doi.org/10.3390/jnt6040034 - 3 Dec 2025
Viewed by 450
Abstract
Obesity and type 2 diabetes are closely linked and often referred to as diabesity. Therapies of diabesity include improving intestinal health and reducing intake of fat and sugars. Diagnosis of diabesity-related metabolic disorders would involve monitoring of glucose and other factors. Nanocellulose, also [...] Read more.
Obesity and type 2 diabetes are closely linked and often referred to as diabesity. Therapies of diabesity include improving intestinal health and reducing intake of fat and sugars. Diagnosis of diabesity-related metabolic disorders would involve monitoring of glucose and other factors. Nanocellulose, also known as cellulose nanomaterials, is emerging as a potential material for various applications. It has unique properties, such as high surface area, biodegradable, biocompatibility and tunable surface chemistry. In this review, we initially provided a brief description of differently produced nanocellulose and their potential applications in different areas, including therapeutics and diagnostics, by focusing on obesity and diabetes. Then, the uptake, absorption, distribution, metabolism and excretion of nanocellulose were discussed. Further, the mechanisms of nanocellulose in modulating diabesity were summarized by emphasizing the role of gut microbiota. Finally, we discussed gut microbiota-related health effects of nanocellulose, both beneficial and detrimental. It was found that the interactions between nanocellulose and gut were complex, with alterations of microbial composition, metabolic activity, and the immune functions both locally and systemically. There seemed to be many beneficial changes following short-term exposure to nanocellulose (e.g., increased beneficial bacteria and decreased pathogenic ones); however, some of these effects were no longer seen after long-term consumption. Importantly, long-term nanocellulose consumption may be associated with certain detrimental health effects, e.g., malnutrition and its associated neurotoxicity, although additional studies are needed to substantiate such health implications. This information is critical for developing safe and effective nanocellulose derivatives that can be applied in food and medicine as well as to harness the benefits of the gut microbiota. Full article
Show Figures

Figure 1

13 pages, 346 KB  
Review
Therapeutic Potential of Leptin in Neurodegenerative Disease
by Jenni Harvey
Biomedicines 2025, 13(12), 2969; https://doi.org/10.3390/biomedicines13122969 - 3 Dec 2025
Viewed by 364
Abstract
Alzheimer’s disease (AD) is an age-related neurodegenerative disorder, characterised by the build-up of amyloid beta (Aβ) plaques and neurofibrillary tangles comprising hyper-phosphorylated tau. Increasing evidence indicates that in the early stages of AD, elevated levels of oligomeric forms of Aβ and phosphorylated tau [...] Read more.
Alzheimer’s disease (AD) is an age-related neurodegenerative disorder, characterised by the build-up of amyloid beta (Aβ) plaques and neurofibrillary tangles comprising hyper-phosphorylated tau. Increasing evidence indicates that in the early stages of AD, elevated levels of oligomeric forms of Aβ and phosphorylated tau (p-tau) gives rise to impaired synaptic function which ultimately drives AD-associated cognitive abnormalities. Thus, developing drugs that can limit the synaptic impairments that occur early in AD may have therapeutic benefits. Clinical evidence increasingly supports a link between lifestyle choices and AD risk. Indeed, there is an association between the circulating levels of the metabolic hormone leptin, mid-life obesity and disease risk, which has in turn stimulated interest in targeting the leptin system to treat AD. It is well-established that leptin readily accesses the brain, with the hippocampus, a key region that degenerates in AD, identified as a prime target for this hormone. Within the hippocampus, leptin has cognitive enhancing properties as it markedly influences the cellular events underlying hippocampal-dependent learning and memory, with significant impact on synaptic plasticity and trafficking of glutamate receptors at hippocampal excitatory CA1 synapses. Moreover, studies using a range of cell-based systems and animal models of disease indicate not only that leptin has powerful pro-cognitive effects, but also that leptin protects against the unwanted synapto-toxic effects of Aβ and tau, as well as enhancing neuronal cell viability. Moreover, recent studies have demonstrated that smaller leptin-based molecules replicate the full repertoire of protective features of whole leptin. Here we review the evidence that the leptin system is a potential novel avenue for drug discovery in AD. Full article
Show Figures

Figure 1

13 pages, 8795 KB  
Brief Report
Safety and Effectiveness of Volumetric Modulated Arc Therapy-Based Stereotactic Radiosurgery for Posterior Fossa Brain Metastases: A Single-Centre Experience
by José Manuel Sánchez-Villalobos, Alfredo Serna-Berna, Juan Salinas-Ramos, Pedro Pablo Escolar-Pérez, Ginés Luengo-Gil, Marina Andreu-Gálvez, Emma Martínez-Alonso and Miguel Alcaraz
J. Clin. Med. 2025, 14(23), 8540; https://doi.org/10.3390/jcm14238540 - 2 Dec 2025
Viewed by 256
Abstract
Background/Objectives: Posterior fossa brain metastases (PFBMs) pose particular risks owing to their proximity to the brainstem and fourth ventricle. We evaluated the safety (treatment-related complications), local effectiveness, and procedural efficiency of volumetric modulated arc therapy (VMAT)-based stereotactic radiosurgery (VMAT-SRS) for PFBMs. Methods: [...] Read more.
Background/Objectives: Posterior fossa brain metastases (PFBMs) pose particular risks owing to their proximity to the brainstem and fourth ventricle. We evaluated the safety (treatment-related complications), local effectiveness, and procedural efficiency of volumetric modulated arc therapy (VMAT)-based stereotactic radiosurgery (VMAT-SRS) for PFBMs. Methods: This single-centre, retrospective study derived a PFBM subgroup from an overall institutional cohort of 123 patients treated with VMAT-RapidArc SRS/fSRS. The doses were 12–20 Gy (single fraction) or 5 × 6 Gy (selected cases). Local response (mRECIST) and predefined safety endpoints (symptomatic oedema with brainstem/IV-ventricle compromise, obstructive hydrocephalus, haemorrhagic transformation, CSF diversion, and urgent neurosurgery) were assessed. Overall survival and procedural time were analysed. Results: Thirty-one patients (39 lesions) were included; 76.9% of them received single-fraction SRS. In addition, 74.2% of patients had supratentorial metastases with posterior fossa involvement. Kaplan–Meier overall survival at 6, 12, 24, and 48 months was 74%, 58%, 26%, and 9.7%, respectively; the median survival time was 12.6 months. Among evaluable lesions, local control was 84.5% (per-lesion response: 15.5% PD, 28.1% SD, 34.4% PR, and 22.0% CR). No clinically significant posterior fossa local complications were observed. Three patients developed radiation-induced leukoencephalopathy after whole-brain radiotherapy (WBRT) and radiosurgery for synchronous supratentorial metastases. The median procedural time was 25.0 min (IQR 9.0) with one isocentre versus 52.5 min (IQR 9.75) with two. Conclusions: VMAT-SRS/fSRS for PFBMs achieved high local control, very low posterior fossa toxicity, and favourable procedural efficiency, supporting its use as a safe, rapid, frameless alternative to WBRT and other radiosurgical platforms such as Gamma Knife in appropriately selected patients. Full article
(This article belongs to the Special Issue New Advances in Stereotactic Radiosurgery)
Show Figures

Figure 1

13 pages, 1633 KB  
Brief Report
The Tetrapeptide HAEE Promotes Amyloid-Beta Clearance from the Brain
by Kristina A. Mukhina, Kseniya B. Varshavskaya, Aleksandra D. Rybak, Viktor V. Grishchenko, Elena V. Kuzubova, Mikhail V. Korokin, Olga I. Kechko and Vladimir A. Mitkevich
Int. J. Mol. Sci. 2025, 26(23), 11591; https://doi.org/10.3390/ijms262311591 - 29 Nov 2025
Viewed by 309
Abstract
Alzheimer’s disease is characterized by the accumulation of neurotoxic forms of amyloid-beta (Aβ) in the brain, leading to synaptic dysfunction, neuroinflammation, and neuronal death. The tetrapeptide HAEE crosses the blood–brain barrier (BBB), inhibits the formation of toxic Aβ oligomers, and reduces amyloid burden [...] Read more.
Alzheimer’s disease is characterized by the accumulation of neurotoxic forms of amyloid-beta (Aβ) in the brain, leading to synaptic dysfunction, neuroinflammation, and neuronal death. The tetrapeptide HAEE crosses the blood–brain barrier (BBB), inhibits the formation of toxic Aβ oligomers, and reduces amyloid burden in vivo. However, the mechanisms of HAEE’s anti-amyloidogenic effect remained incompletely understood. In this study, we investigated the mechanism of HAEE-dependent Aβ clearance both in vitro and in vivo. Using ELISA, we assessed the HAEE effect on the levels of Aβ, IL-6, and TNFα in mouse brain tissue following intracerebroventricular administration. The mechanism of the anti-Aβ effect of HAEE was studied using primary brain cell cultures and a BBB transwell model through ELISA, flow cytometry, and microscopy. We showed that HAEE reduced Aβ level by 35% and IL-6 level by 40% in mouse brain tissue. HAEE enhanced Aβ clearance via LRP1- and PgP-dependent Aβ transport through the BBB and doubled the rate of Aβ degradation by microglia. In addition to inhibition of Aβ aggregation, HAEE dissolved already formed Aβ oligomers. The HAEE-induced decrease in IL-6 levels in the mouse brain was associated with reduced pro-inflammatory activation of microglia. Thus, HAEE’s effect against Aβ-related neuropathologies is realized through a decrease in the level of toxic Aβ oligomer and inhibition of neuroinflammation. Full article
Show Figures

Figure 1

15 pages, 1439 KB  
Article
Resveratrol Mitigates High Glucose-Induced Inflammation in Astroglial Cells
by Vanessa Sovrani, Filipe Renato Pereira Dias, Rômulo Rodrigo de Souza Almeida, Krista Minéia Wartchow, Nícolas Manzke Glänzel, Ester Rezena, Carlos-Alberto Gonçalves, Guilhian Leipnitz, Larissa Daniele Bobermin and André Quincozes-Santos
Metabolites 2025, 15(12), 771; https://doi.org/10.3390/metabo15120771 - 28 Nov 2025
Viewed by 330
Abstract
Background/Objectives: Changes in glucose metabolism impact central nervous system (CNS) homeostasis and, consequently, can lead to cognitive impairment and an increased risk for neurodegenerative and neuropsychiatric disorders. Astrocytes are glial cells that act as key regulators of brain glucose metabolism, thus representing important [...] Read more.
Background/Objectives: Changes in glucose metabolism impact central nervous system (CNS) homeostasis and, consequently, can lead to cognitive impairment and an increased risk for neurodegenerative and neuropsychiatric disorders. Astrocytes are glial cells that act as key regulators of brain glucose metabolism, thus representing important cellular targets for studies of different pathophysiological conditions, including hyperglycemia. Resveratrol, a natural polyphenol, has emerged as a potential protective strategy against diabetes and its complications; however, its glioprotective effects remain unclear. Based on these observations, we evaluated whether resveratrol could modify the inflammatory response in astroglial cells exposed to experimental hyperglycemic conditions. Methods: After reaching confluence, C6 astroglial cells were pre-incubated with 10 µM resveratrol in serum-free DMEM with 6 mM glucose for 24 h. The medium was then replaced with serum-free DMEM containing 12 mM glucose and 10 µM resveratrol for another 24 h. Controls were maintained in 6 mM glucose. Analyses included cell viability, metabolic activity, glucose and glutamate uptake, cytokine quantification by ELISA, and gene expression by RT-qPCR. Results: We show that high glucose levels modulate glucose and glutamate metabolism, and increase neuroinflammation, through the modulation of inflammatory mediators. In addition, high glucose upregulated the gene expressions of inducible nitric oxide synthase (iNOS), nuclear factor κB (NFκB), cyclooxygenase 2 (COX2), and Toll-like receptor 4 (TLR4) while decreasing mRNA levels of NLR family pyrin domain containing 3 (NLRP3) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). However, resveratrol was able to prevent most of these effects, particularly the high glucose-triggered inflammatory response. Resveratrol also modulated heme oxygenase 1 (HO-1) and nuclear factor erythroid-derived 2-like 2 (Nrf2), important targets associated with cellular protection. Conclusions: Our findings reinforce resveratrol as a potential glioprotective strategy against diabetes-related brain toxicity. Full article
(This article belongs to the Special Issue Metabolic Profiling in Neurometabolisms)
Show Figures

Figure 1

Back to TopTop