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
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 (4,051)

Search Parameters:
Keywords = brain modules

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 1875 KiB  
Review
Translating Exosomal microRNAs from Bench to Bedside in Parkinson’s Disease
by Oscar Arias-Carrión, María Paulina Reyes-Mata, Joaquín Zúñiga and Daniel Ortuño-Sahagún
Brain Sci. 2025, 15(7), 756; https://doi.org/10.3390/brainsci15070756 - 16 Jul 2025
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder marked by dopaminergic neuronal loss, α-synuclein aggregation, and chronic neuroinflammation. Recent evidence suggests that exosomal microRNAs (miRNAs)—small, non-coding RNAs encapsulated in extracellular vesicles—are key regulators of PD pathophysiology and promising candidates for biomarker development and [...] Read more.
Parkinson’s disease (PD) is a progressive neurodegenerative disorder marked by dopaminergic neuronal loss, α-synuclein aggregation, and chronic neuroinflammation. Recent evidence suggests that exosomal microRNAs (miRNAs)—small, non-coding RNAs encapsulated in extracellular vesicles—are key regulators of PD pathophysiology and promising candidates for biomarker development and therapeutic intervention. Exosomes facilitate intercellular communication, cross the blood–brain barrier, and protect miRNAs from degradation, rendering them suitable for non-invasive diagnostics and targeted delivery. Specific exosomal miRNAs modulate neuroinflammatory cascades, oxidative stress, and synaptic dysfunction, and their altered expression in cerebrospinal fluid and plasma correlates with disease onset, severity, and progression. Despite their translational promise, challenges persist, including methodological variability in exosome isolation, miRNA profiling, and delivery strategies. This review integrates findings from preclinical models, patient-derived samples, and systems biology to delineate the functional impact of exosomal miRNAs in PD. We propose mechanistic hypotheses linking miRNA dysregulation to molecular pathogenesis and present an interactome model highlighting therapeutic nodes. Advancing exosomal miRNA research may transform the clinical management of PD by enabling earlier diagnosis, molecular stratification, and the development of disease-modifying therapies. Full article
(This article belongs to the Special Issue Molecular Insights in Neurodegeneration)
Show Figures

Figure 1

21 pages, 2793 KiB  
Article
Link Predictions with Bi-Level Routing Attention
by Yu Wang, Shu Xu, Zenghui Ding, Cong Liu and Xianjun Yang
AI 2025, 6(7), 156; https://doi.org/10.3390/ai6070156 - 14 Jul 2025
Viewed by 127
Abstract
Background/Objectives: Knowledge Graphs (KGs) are often incomplete, which can significantly impact the performance of downstream applications. Manual completion of KGs is time-consuming and costly, emphasizing the importance of developing automated methods for KGC. Link prediction serves as a fundamental task in this domain. [...] Read more.
Background/Objectives: Knowledge Graphs (KGs) are often incomplete, which can significantly impact the performance of downstream applications. Manual completion of KGs is time-consuming and costly, emphasizing the importance of developing automated methods for KGC. Link prediction serves as a fundamental task in this domain. The semantic correlation among entity features plays a crucial role in determining the effectiveness of link-prediction models. Notably, the human brain can often infer information using a limited set of salient features. Methods: Inspired by this cognitive principle, this paper proposes a lightweight Bi-level routing attention mechanism specifically designed for link-prediction tasks. This proposed module explores a theoretically grounded and lightweight structural design aimed at enhancing the semantic recognition capability of language models without altering their core parameters. The proposed module enhances the model’s ability to attend to feature regions with high semantic relevance. With only a marginal increase of approximately one million parameters, the mechanism effectively captures the most semantically informative features. Result: It replaces the original feature-extraction module within the KGML framework and is evaluated on the publicly available WN18RR and FB15K-237 dataset. Conclusions: Experimental results demonstrate consistent improvements in standard evaluation metrics, including Mean Rank (MR), Mean Reciprocal Rank (MRR), and Hits@10, thereby confirming the effectiveness of the proposed approach. Full article
Show Figures

Figure 1

36 pages, 914 KiB  
Review
Gut Microbiota in Women with Eating Disorders: A New Frontier in Pathophysiology and Treatment
by Giuseppe Marano, Sara Rossi, Greta Sfratta, Mariateresa Acanfora, Maria Benedetta Anesini, Gianandrea Traversi, Francesco Maria Lisci, Lucio Rinaldi, Roberto Pola, Antonio Gasbarrini, Gabriele Sani, Eleonora Gaetani and Marianna Mazza
Nutrients 2025, 17(14), 2316; https://doi.org/10.3390/nu17142316 - 14 Jul 2025
Viewed by 146
Abstract
Emerging evidence highlights the critical role of the gut microbiota in the development and progression of eating disorders (EDs), particularly in women, who are more frequently affected by these conditions. Women with anorexia nervosa, bulimia nervosa, and binge eating disorder exhibit distinct alterations [...] Read more.
Emerging evidence highlights the critical role of the gut microbiota in the development and progression of eating disorders (EDs), particularly in women, who are more frequently affected by these conditions. Women with anorexia nervosa, bulimia nervosa, and binge eating disorder exhibit distinct alterations in gut microbiota composition compared to healthy controls. These alterations, collectively termed dysbiosis, involve reduced microbial diversity and shifts in key bacterial populations responsible for regulating metabolism, inflammation, and gut–brain signaling. The gut microbiota is known to influence appetite regulation, mood, and stress responses—factors closely implicated in the pathogenesis of EDs. In women, hormonal fluctuations related to menstruation, pregnancy, and menopause may further modulate gut microbial profiles, potentially compounding vulnerabilities to disordered eating. Moreover, the restrictive eating patterns, purging behaviors, and altered dietary intake often observed in women with EDs exacerbate microbial imbalances, contributing to intestinal permeability, low-grade inflammation, and disturbances in neurotransmitter production. This evolving understanding suggests that microbiota-targeted therapies, such as probiotics, prebiotics, dietary modulation, and fecal microbiota transplantation (FMT), could complement conventional psychological and pharmacological treatments in women with EDs. Furthermore, precision nutrition and personalized microbiome-based interventions tailored to an individual’s microbial and metabolic profile offer promising avenues for improving treatment efficacy, even though these approaches remain exploratory and their clinical applicability has yet to be fully validated. Future research should focus on sex-specific microbial signatures, causal mechanisms, and microbiota-based interventions to enhance personalized treatment for women struggling with eating disorders. Full article
(This article belongs to the Section Clinical Nutrition)
Show Figures

Figure 1

12 pages, 1732 KiB  
Article
EEG-Based Analysis of Neural Responses to Sweeteners: Effects of Type and Concentration
by Xiaolei Wang, Guangnan Wang and Donghong Liu
Foods 2025, 14(14), 2460; https://doi.org/10.3390/foods14142460 - 14 Jul 2025
Viewed by 163
Abstract
Sweetness is a key dimension of sensory experience in food, and variations in the type and concentration of sweeteners can elicit distinct brain responses. In this study, electroencephalography (EEG) was employed to systematically evaluate neural activity elicited by different concentrations of sucrose solutions [...] Read more.
Sweetness is a key dimension of sensory experience in food, and variations in the type and concentration of sweeteners can elicit distinct brain responses. In this study, electroencephalography (EEG) was employed to systematically evaluate neural activity elicited by different concentrations of sucrose solutions (1%, 3%, 5%, and 7%) and by non-nutritive sweeteners matched in perceived sweetness to a 7% sucrose solution (10% erythritol, 0.0133% sucralose, and 0.0368% stevioside). The results revealed that an increased sucrose concentration was associated with progressively weaker EEG signal intensity, suggesting that the brain can effectively distinguish sweetness intensity. Under iso-sweet conditions, different types of sweeteners induced significantly distinct EEG patterns, indicating that the nature of the sweetener modulates flavor perception at the neural level. Further analysis showed increases in both δ- and α-band power following sweet taste stimulation, with prominent activations observed in the frontal, parietal, and right temporal regions. These findings demonstrate the utility of EEG in detecting subtle differences in brain responses to sweeteners, offering new insights into the neural mechanisms underlying sweet taste perception. Full article
(This article belongs to the Section Sensory and Consumer Sciences)
Show Figures

Figure 1

11 pages, 522 KiB  
Review
The Role of Gut Microbiota in Suicidality: Mechanisms, Evidence, and Future Directions
by Valentina Baldini, Martina Gnazzo, Giulia Santangelo, Giorgia Varallo, Diana De Ronchi and Marco Carotenuto
Psychiatry Int. 2025, 6(3), 84; https://doi.org/10.3390/psychiatryint6030084 - 14 Jul 2025
Viewed by 111
Abstract
Suicidality, encompassing suicidal ideation, attempts, and completed suicide, continues to be a significant public health concern globally. Traditional research has emphasized genetic, neurobiological, and psychosocial factors; however, recent findings suggest that gut microbiota may play a crucial role in influencing suicidal behavior. The [...] Read more.
Suicidality, encompassing suicidal ideation, attempts, and completed suicide, continues to be a significant public health concern globally. Traditional research has emphasized genetic, neurobiological, and psychosocial factors; however, recent findings suggest that gut microbiota may play a crucial role in influencing suicidal behavior. The gut microbiota impacts neuroinflammation, neurotransmitter metabolism, and the hypothalamic–pituitary–adrenal (HPA) axis, all of which are associated with psychiatric disorders linked to suicidality. This review gathers current evidence on the gut–brain axis, investigating the role of microbiota in suicidality through mechanisms such as immune system modulation, serotonin regulation, and the stress response. We also consider the potential of microbiota-targeted interventions, such as probiotics and dietary changes, as innovative therapeutic strategies. Despite the accumulating evidence, research in this field remains limited, emphasizing the urgent need for further investigation to clarify the causal relationship between gut microbiota and suicidality. Full article
Show Figures

Figure 1

29 pages, 4862 KiB  
Article
Repurposed Antipsychotics as Potential Anticancer Agents: Clozapine Efficacy and Dopaminergic Pathways in Neuroblastoma and Glioblastoma
by Catarina Moura, Maria João Gouveia and Nuno Vale
Life 2025, 15(7), 1097; https://doi.org/10.3390/life15071097 - 12 Jul 2025
Viewed by 219
Abstract
Neuro-oncology focuses on the diagnosis and treatment of brain tumors, which, despite their rarity, are associated with high mortality due to their invasiveness and limited treatment options. Emerging evidence suggests that dopamine (DA), a neurotransmitter crucial for cognitive and emotional processes, and its [...] Read more.
Neuro-oncology focuses on the diagnosis and treatment of brain tumors, which, despite their rarity, are associated with high mortality due to their invasiveness and limited treatment options. Emerging evidence suggests that dopamine (DA), a neurotransmitter crucial for cognitive and emotional processes, and its receptors may influence tumor growth and the tumor microenvironment. This study aimed to evaluate the potential anticancer effects of repurposed antipsychotic dopamine-targeting drugs (Clozapine, CLZ; Pimozide, PIM; Olanzapine, OLZ; and Risperidone, RIS) and antiemetic drugs (Domperidone, DOM; Droperidol, DRO) on neuroblastoma (SH-SY5Y) and glioblastoma (A172) cell lines, and to assess whether their efficacy is modulated by oxidative stress and DA synthesis. The drugs were first tested individually, followed by co-treatment with tyrosine (Tyr), a dopamine precursor, and hydrogen peroxide (H2O2), an inducer of oxidative stress. Additionally, drug activity was evaluated in the simultaneous presence of H2O2 and Tyr. CLZ exhibited the highest cytotoxicity in both cell lines, suggesting strong anticancer potential and also synergism among the different combinations, particularly in SH-SY5Y. Liquid chromatography of the extracellular medium showed greater Tyr consumption in SH-SY5Y compared to A172 cells, indicating a higher dependence on extracellular Tyr to mitigate drug- and/or stress-induced cytotoxicity. In summary, several of the repurposed antipsychotics demonstrated cytotoxic effects on central nervous system tumor cells, with CLZ showing the most promising activity, even under oxidative stress conditions. These findings support further investigation into dopamine-targeting drugs as potential therapeutic agents in neuro-oncology. Full article
(This article belongs to the Section Pharmaceutical Science)
Show Figures

Figure 1

17 pages, 7114 KiB  
Article
Synthetic Feed Attractants in European Seabass (Dicentrarchus labrax) Culture: Effects on Growth, Health, and Appetite Stimulation
by Federico Conti, Matteo Zarantoniello, Nico Cattaneo, Matteo Antonucci, Elena Antonia Belfiore and Ike Olivotto
Animals 2025, 15(14), 2060; https://doi.org/10.3390/ani15142060 - 12 Jul 2025
Viewed by 368
Abstract
Synthetic flavors from standardized processes have recently emerged as a promising and sustainable alternative to traditional feed attractants. In this study, two attractive (F25, cheese; F35, caramel) and one repulsive (F32-, coconut) synthetic flavors were individually added (1% w/w) to [...] Read more.
Synthetic flavors from standardized processes have recently emerged as a promising and sustainable alternative to traditional feed attractants. In this study, two attractive (F25, cheese; F35, caramel) and one repulsive (F32-, coconut) synthetic flavors were individually added (1% w/w) to a commercial diet for European seabass (Dicentrarchus labrax) and tested over a 90-day feeding trial (30 fish per tank, in triplicate; initial weight 72.48 ± 8.04 g) to assess their impact on fish growth performance, welfare, and the modulation of brain appetite and monoaminergic pathways. None of the tested flavors negatively affected overall fish health. The F35 flavor enhanced feed intake (90.1 ± 5.6%) and growth (SGR 2.2 ± 0.2%) and positively influenced appetite-related and monoaminergic signals, thus being more effective than the F25 one (80.4 ± 3.2 and 1.6 ± 0.1%, respectively). A weekly feeding rotation between F35 and F25 (ROT group) resulted in suboptimal outcomes compared to F35 administration alone. The F32- flavor was not clearly perceived as strongly aversive by seabass and did not impair zootechnical performance. These findings highlight the potential of attractive synthetic flavors to improve diet palatability in a carnivorous species of commercial value, offering novel insights for more sustainable and cost-effective aquaculture feeding strategies. Full article
(This article belongs to the Special Issue Recent Advances in Nutritional Ingredients for Aquaculture)
Show Figures

Figure 1

36 pages, 1414 KiB  
Review
A Systems Biology Approach to Memory Health: Integrating Network Pharmacology, Gut Microbiota, and Multi-Omics for Health Functional Foods
by Heng Yuan, Junyu Zhou, Hongbao Li, Suna Kang and Sunmin Park
Int. J. Mol. Sci. 2025, 26(14), 6698; https://doi.org/10.3390/ijms26146698 - 12 Jul 2025
Viewed by 154
Abstract
Memory impairment, ranging from mild memory impairment to neurodegenerative diseases such as Alzheimer’s disease, poses an escalating global health challenge that necessitates multi-targeted interventions to prevent progression. Health functional foods (HFFs), which include bioactive dietary compounds that not only provide basic nutrition but [...] Read more.
Memory impairment, ranging from mild memory impairment to neurodegenerative diseases such as Alzheimer’s disease, poses an escalating global health challenge that necessitates multi-targeted interventions to prevent progression. Health functional foods (HFFs), which include bioactive dietary compounds that not only provide basic nutrition but also function beyond that to modulate physiological pathways, offer a promising non-pharmacological strategy to preserve memory function. This review presents an integrative framework for the discovery, evaluation, and clinical translation of biomarkers responsive to HFFs in the context of preventing memory impairment. We examine both established clinical biomarkers, such as amyloid-β and tau in the cerebrospinal fluid, neuroimaging indicators, and memory assessments, as well as emerging nutritionally sensitive markers including cytokines, microRNAs, gut microbiota signatures, epigenetic modifications, and neuroactive metabolites. By leveraging systems biology approaches, we explore how network pharmacology, gut–brain axis modulation, and multi-omics integration can help to elucidate the complex interactions between HFF components and memory-related pathways such as neuroinflammation, oxidative stress, synaptic plasticity, and metabolic regulation. The review also addresses the translational pipeline for HFFs, from formulation and standardization to regulatory frameworks and clinical development, with an emphasis on precision nutrition strategies and cross-disciplinary integration. Ultimately, we propose a paradigm shift in memory health interventions, positioning HFFs as scientifically validated compounds for personalized nutrition within a preventative memory function framework. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Alzheimer’s Disease)
Show Figures

Figure 1

22 pages, 637 KiB  
Article
Congee Containing Carotenoids-Enriched Functional Ingredient from Tomato Improves Cognition, Serum α-Synuclein, Monoaminergic Function, and Gut-Brain Axis Functions in the Elderly Volunteers
by Jintanaporn Wattanathorn, Supaporn Muchimapura, Wipawee Thukham-mee and Terdthai Tong-un
Life 2025, 15(7), 1093; https://doi.org/10.3390/life15071093 - 11 Jul 2025
Viewed by 208
Abstract
Owing to the need for a brain supplement targeting neuroprotection against age-related brain disorders and the known effect of carotenoids on brain function, we aimed to assess the effect of consuming carotenoid-rich functional congee for 8 weeks on cognitive function and age-related serum [...] Read more.
Owing to the need for a brain supplement targeting neuroprotection against age-related brain disorders and the known effect of carotenoids on brain function, we aimed to assess the effect of consuming carotenoid-rich functional congee for 8 weeks on cognitive function and age-related serum biomarkers. Both males and females (60–70 years old) were randomly assigned to consume either placebo or carotenoid-rich functional congee containing dried tomato powder at doses of 200 and 400 mg per day. Cognitive function, working memory, and serum biomarkers including alpha-synuclein and PARK7, together with serum oxidative stress parameters and neurotransmitters, were assessed prior to consumption and every 4 weeks throughout the study period. The contents of Lactobacillus and Bifidobacterium spp. in feces were also determined. Both doses of carotenoid-enriched congee enhanced cognitive function (P300), but only low doses improved working memory and decreased the activity of MAO-A and serum alpha-synuclein. The high-dose congee-treated group exhibited an increase in the density of Lactobacillus spp. in feces. Taking all data together, the carotenoid-enriched congee enhances cognitive function and working memory. The mechanisms may partly involve the increase in monoaminergic function, while the modulation of the gut–brain axis may require further confirmation. Full article
Show Figures

Figure 1

21 pages, 27301 KiB  
Article
Folic Acid Ameliorates Neuronal Ferroptosis in Aging by Up-Regulating SLC7A11-GSH-GPX4 Antioxidant Pathway and Increasing Cystine Levels
by Yue Wang, Jingwen Zhang, Zehao Wang, Qinghan Ren, Zhenshu Li, Guowei Huang and Wen Li
Int. J. Mol. Sci. 2025, 26(14), 6669; https://doi.org/10.3390/ijms26146669 - 11 Jul 2025
Viewed by 185
Abstract
Age-related neurodegeneration is characterized by oxidative stress and iron-dependent cell death, yet the neuroprotective mechanisms of folic acid in modulating ferroptosis remain unclear. This study systematically investigated the role of folic acid in inhibiting ferroptosis and attenuating neuronal damage in aging, with a [...] Read more.
Age-related neurodegeneration is characterized by oxidative stress and iron-dependent cell death, yet the neuroprotective mechanisms of folic acid in modulating ferroptosis remain unclear. This study systematically investigated the role of folic acid in inhibiting ferroptosis and attenuating neuronal damage in aging, with a focus on the solute carrier family 7 member 11 (SLC7A11)-glutathione (GSH)-glutathione peroxidase 4 (GPX4) antioxidant pathway, using aged rats supplemented with folic acid (<0.1, 2.0, and 4.0 mg/kg·diet) for 22 months, with young adult rats as controls. Brain iron accumulation and ferroptosis-related proteins (SLC7A11, GPX4, Ferritin heavy chain 1 (FTH1)) were evaluated. In vitro, HT-22 hippocampal neuronal cells were pre-treated with folic acid (0, 10, 20 μmol/L) for 72 h before combining with Erastin (10 μmol/L)-induced ferroptosis for an additional 24 h. Intracellular Fe2+, lipid peroxidation (LPO), malondialdehyde (MDA), reactive oxygen species (ROS), along with cystine, GSH, and ferroptosis-related protein levels were quantified. Stable sh-SLC7A11 knockdown and control (sh-NC) cell lines were used to validate the dependency of folic acid’s protective effects on SLC7A11 expression. Folic acid supplementation in aged rats dose-dependently reduced aging-related brain iron accumulation and enhanced the expression of SLC7A11, GPX4, and FTH1. In Erastin-induced HT-22 cells, folic acid significantly mitigated ferroptosis hallmarks. Mechanistically, folic acid increased extracellular cystine uptake and intracellular GSH synthesis, thereby activating the SLC7A11-GSH-GPX4 antioxidant pathway. Notably, molecular docking technique suggested that compared to GPX4, folic acid stabilized SLC7A11’s active conformation. sh-SLC7A11 knockdown completely abolished folic acid-mediated protection against ferroptosis, as evidenced by restored loss of cystine, GSH and GPX4 production. This study innovatively emphasized the critical role of folic acid supplementation in inhibiting ferroptosis by up-regulating the SLC7A11-GSH-GPX4 antioxidant pathway, primarily through enhancing cystine availability and SLC7A11 expression. These findings established folic acid as a potential dietary intervention for aging-related neurodegenerative diseases characterized by neuronal ferroptosis, providing preclinical evidence for folic acid based neuroprotection. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

29 pages, 17950 KiB  
Article
Organ-Specific Small Protein Networks in 100 kDa Ultrafiltrates: Functional Analysis and Implications for Neuroregenerative Medicine
by Jakub Peter Slivka, Chris Bauer, Tasneem Halhouli, Alexander Younsi, Michelle B. F. Wong, Mike K. S. Chan and Thomas Skutella
Int. J. Mol. Sci. 2025, 26(14), 6659; https://doi.org/10.3390/ijms26146659 - 11 Jul 2025
Viewed by 117
Abstract
In this research, the proteomic landscape of 100 kDa protein extract sourced from rabbit brain was compared to extracts from liver and from organ mixture (OM). Our aim was to compare the efficacy of Nanomised Organo Peptides (NOP) ultrafiltrates from two different tissues [...] Read more.
In this research, the proteomic landscape of 100 kDa protein extract sourced from rabbit brain was compared to extracts from liver and from organ mixture (OM). Our aim was to compare the efficacy of Nanomised Organo Peptides (NOP) ultrafiltrates from two different tissues and a tissue mixture for inducing neurite outgrowth, and subsequently to identify the molecular networks and proteins that could explain such effects. Proteins were isolated by gentle homogenization followed by crossflow ultrafiltration. Proteomic evaluation involved gel electrophoresis, complemented by mass spectrometry and bioinformatics. GO (Gene Ontology) and protein analysis of the mass spectrometry results identified a diverse array of proteins involved in critical specific biological functions, including neuronal development, regulation of growth, immune response, and lipid and metal binding. Data from this study are accessible from the ProteomeXchange repository (identifier PXD051701). Our findings highlight the presence of small proteins that play key roles in metabolic processes and biosynthetic modulation. In vitro outgrowth experiments with neural stem cells (NSCs) showed that 100 kDa protein extracts from the brain resulted in a greater increase in neurite length compared to the liver and organ mixture extracts. The protein networks identified in the NOP ultrafiltrates may significantly improve biological therapeutic strategies related to neural differentiation and outgrowth. This comprehensive proteomic analysis of 100 kDa ultrafiltrates revealed a diverse array of proteins involved in key biological processes, such as neuronal development, metabolic regulation, and immune response. Brain-specific extracts demonstrated the capacity to promote neurite outgrowth in NSCs, suggesting potential application for neuroregenerative therapies. Our findings highlight the potential of small proteins and organ-specific proteins in the development of novel targeted treatments for various diseases, particularly those related to neurodegeneration and aging. Full article
Show Figures

Figure 1

40 pages, 1203 KiB  
Review
Overview of Preclinical and Clinical Trials of Nanoparticles for the Treatment of Brain Metastases
by Muhammad Izhar, Mohamed Al Gharyani, Ahed H. Kattaa, Juan J. Cardona, Ruchit P. Jain, Elaheh Shaghaghian, Yusuke S. Hori, Fred C. Lam, Deyaaldeen Abu Reesh, Sara C. Emrich, Louisa Ustrzynski, Armine Tayag, Maciej S. Lesniak, Steven D. Chang and David J. Park
Pharmaceutics 2025, 17(7), 899; https://doi.org/10.3390/pharmaceutics17070899 - 11 Jul 2025
Viewed by 328
Abstract
Brain metastases (BM), which most commonly originate from lung, breast, or skin cancers, remain a major clinical challenge, with standard treatments such as stereotactic radiosurgery (SRS), surgical resection, and whole-brain radiation therapy (WBRT). The prognosis for patients with BM remains poor, with a [...] Read more.
Brain metastases (BM), which most commonly originate from lung, breast, or skin cancers, remain a major clinical challenge, with standard treatments such as stereotactic radiosurgery (SRS), surgical resection, and whole-brain radiation therapy (WBRT). The prognosis for patients with BM remains poor, with a median overall survival (OS) of just 10–16 months. Although recent advances in systemic therapies, including small molecule inhibitors, monoclonal antibodies, chemotherapeutics, and gene therapies, have demonstrated success in other malignancies, their effectiveness in central nervous system (CNS) cancers is significantly limited by poor blood–brain barrier (BBB) permeability and subtherapeutic drug concentrations in the brain. Nanoparticle-based drug delivery systems have emerged as a promising strategy to overcome these limitations by enhancing CNS drug penetration and selectively targeting metastatic brain tumor cells while minimizing off-target effects. This review summarizes recent preclinical and clinical developments in nanoparticle-based therapies for BM. It is evident from these studies that NPs can carry with them a range of therapeutics, including chemotherapy, immunotherapy, small molecule inhibitors, gene therapies, radiosensitizers, and modulators of tumor microenvironment to the BM. Moreover, preclinical studies have shown encouraging efficacy in murine models, highlighting the potential of these platforms to improve therapeutic outcomes. However, clinical translation remains limited, with few ongoing trials. To close this translational gap, future work must address clinical challenges such as trial design, regulatory hurdles, and variability in BBB permeability while developing personalized nanoparticle-based therapies tailored to individual tumor characteristics. Full article
(This article belongs to the Special Issue Development of Novel Tumor-Targeting Nanoparticles, 2nd Edition)
Show Figures

Figure 1

30 pages, 34072 KiB  
Article
ARE-PaLED: Augmented Reality-Enhanced Patch-Level Explainable Deep Learning System for Alzheimer’s Disease Diagnosis from 3D Brain sMRI
by Chitrakala S and Bharathi U
Symmetry 2025, 17(7), 1108; https://doi.org/10.3390/sym17071108 - 10 Jul 2025
Viewed by 284
Abstract
Structural magnetic resonance imaging (sMRI) is a vital tool for diagnosing neurological brain diseases. However, sMRI scans often show significant structural changes only in limited brain regions due to localised atrophy, making the identification of discriminative features a key challenge. Importantly, the human [...] Read more.
Structural magnetic resonance imaging (sMRI) is a vital tool for diagnosing neurological brain diseases. However, sMRI scans often show significant structural changes only in limited brain regions due to localised atrophy, making the identification of discriminative features a key challenge. Importantly, the human brain exhibits inherent bilateral symmetry, and deviations from this symmetry—such as asymmetric atrophy—are strong indicators of early Alzheimer’s disease (AD). Patch-based methods help capture local brain changes for early AD diagnosis, but they often struggle with fixed-size limitations, potentially missing subtle asymmetries or broader contextual cues. To address these limitations, we propose a novel augmented reality (AR)-enhanced patch-level explainable deep learning (ARE-PaLED) system. It includes an adaptive multi-scale patch extraction network (AMPEN) to adjust patch sizes based on anatomical characteristics and spatial context, as well as an informative patch selection algorithm (IPSA) to identify discriminative patches, including those reflecting asymmetry patterns associated with AD; additionally, an AR module is proposed for future immersive explainability, complementing the patch-level interpretation framework. Evaluated on 1862 subjects from the ADNI and AIBL datasets, the framework achieved an accuracy of 92.5% (AD vs. NC) and 85.9% (AD vs. MCI). The proposed ARE-PaLED demonstrates potential as an interpretable and immersive diagnostic aid for sMRI-based AD diagnosis, supporting the interpretation of model predictions for AD diagnosis. Full article
Show Figures

Figure 1

25 pages, 1614 KiB  
Review
Intermittent Fasting as a Neuroprotective Strategy: Gut–Brain Axis Modulation and Metabolic Reprogramming in Neurodegenerative Disorders
by Zaw Myo Hein, Muhammad Faqhrul Fahmy Arbain, Suresh Kumar, Muhammad Zulfadli Mehat, Hafizah Abdul Hamid, Muhammad Danial Che Ramli and Che Mohd Nasril Che Mohd Nassir
Nutrients 2025, 17(14), 2266; https://doi.org/10.3390/nu17142266 - 9 Jul 2025
Viewed by 658
Abstract
Intermittent fasting (IF) is emerging as a heterogeneous neurometabolic intervention with the possibility of changing the course of neurodegenerative diseases. Through the modulation of the gut–brain axis (GBA), cellular bioenergetics (or metabolic) reprogramming, and involvement in preserved stress adaptation pathways, IF influences a [...] Read more.
Intermittent fasting (IF) is emerging as a heterogeneous neurometabolic intervention with the possibility of changing the course of neurodegenerative diseases. Through the modulation of the gut–brain axis (GBA), cellular bioenergetics (or metabolic) reprogramming, and involvement in preserved stress adaptation pathways, IF influences a range of physiological mechanisms, including mitobiogenesis, autophagy, circadian rhythm alignment, and neuroinflammation. This review critically synthesises current preclinical and early clinical evidence illustrating IF’s capability to supplement synaptic plasticity and integrity, reduce toxic proteins (proteotoxic) burden, and rehabilitate glial and immune homeostasis across models of Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. The key players behind these effects are bioactive metabolites such as short-chain fatty acids (SCFA) and β-hydroxybutyrate (BHB), and molecular mediators such as brain-derived neurotrophic factor (BDNF). We feature the therapeutic pertinence of IF-induced changes in gut microbiota composition, immune response, and mitochondrial dynamics, and we discuss emerging approaches for merging IF into precision medicine frameworks. Crucial challenges include individual variability, protocol optimisation, safety in cognitively vulnerable populations, and the need for biomarker-guided, ethically grounded clinical trials. Finally, we propose IF as a scalable and flexible intervention that, when personalised and integrated with other modalities, may reframe neurodegeneration from a model of irreversible decline to one of modifiable resilience. Full article
(This article belongs to the Section Nutrition and Neuro Sciences)
Show Figures

Figure 1

25 pages, 2464 KiB  
Systematic Review
Modulating the Gut Microbiota to Target Neuroinflammation, Cognition and Mood: A Systematic Review of Human Studies with Relevance to Fibromyalgia
by Gianna Dipalma, Grazia Marinelli, Laura Ferrante, Angela Di Noia, Claudio Carone, Valeria Colonna, Pierluigi Marotti, Francesco Inchingolo, Andrea Palermo, Gianluca Martino Tartaglia, Massimo Del Fabbro, Angelo Michele Inchingolo and Alessio Danilo Inchingolo
Nutrients 2025, 17(14), 2261; https://doi.org/10.3390/nu17142261 - 9 Jul 2025
Viewed by 403
Abstract
Aim: This systematic review aims to evaluate the effectiveness of microbiota-modulating interventions (such as probiotics, prebiotics, and fecal microbiota transplantation) in reducing cognitive symptoms, pain, and neuroinflammation in human studies relevant to fibromyalgia (FM). The review will investigate the role of gut–brain axis [...] Read more.
Aim: This systematic review aims to evaluate the effectiveness of microbiota-modulating interventions (such as probiotics, prebiotics, and fecal microbiota transplantation) in reducing cognitive symptoms, pain, and neuroinflammation in human studies relevant to fibromyalgia (FM). The review will investigate the role of gut–brain axis modulation through these interventions and explore the potential therapeutic benefits for FM management. Materials and Methods: A comprehensive search was conducted in electronic databases including PubMed, Scopus, and the Cochrane Library for studies published from 1 January 2015 to 30 April 2025. Studies were eligible if they were randomized controlled trials (RCTs), pilot studies, or observational studies assessing the impact of microbiota-targeted interventions (probiotics, prebiotics, fecal microbiota transplantation) on cognitive function, pain, or neuroinflammation in patients with FM. Studies were excluded if they involved animal models, lacked relevant outcome measures, or were not peer-reviewed. Although only a subset of the included studies directly involved FM patients, all were selected for their relevance to symptom domains (e.g., pain, cognition, mood) and mechanisms (e.g., neuroinflammation, gut–brain axis dysfunction) that are central to FM. A total of 11 human studies were included in the final qualitative synthesis. Results: Preliminary findings from the included studies suggest that microbiota-targeted interventions, particularly probiotics and prebiotics, show promise in reducing cognitive symptoms, pain, and neuroinflammation in FM patients. Improvements in mood and quality of life were also reported, indicating potential benefits for overall well-being. However, heterogeneity in study designs, sample sizes, and outcome measures limit the ability to draw definitive conclusions. Conclusions: This systematic review highlights the potential of microbiota modulation as a therapeutic strategy for managing FM symptoms, particularly cognitive dysfunction and neuroinflammation. Full article
(This article belongs to the Special Issue Implications of Diet and the Gut Microbiome in Neuroinflammation)
Show Figures

Figure 1

Back to TopTop