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
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
remove_circle_outline

Search Results (2,800)

Search Parameters:
Keywords = metabolic dysregulation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 934 KB  
Review
Antioxidant Effect of Curcumin and Its Impact on Mitochondria: Evidence from Biological Models
by Karla Alejandra Avendaño-Briseño, Jorge Escutia-Martínez, Estefani Yaquelin Hernández-Cruz and José Pedraza-Chaverri
J. Xenobiot. 2025, 15(5), 139; https://doi.org/10.3390/jox15050139 (registering DOI) - 31 Aug 2025
Abstract
Curcumin, the principal active component of turmeric, is a polyphenol that has been used in various countries for the treatment of numerous conditions due to its wide range of health benefits. Curcumin exhibits bifunctional antioxidant properties: the first is attributed to its chemical [...] Read more.
Curcumin, the principal active component of turmeric, is a polyphenol that has been used in various countries for the treatment of numerous conditions due to its wide range of health benefits. Curcumin exhibits bifunctional antioxidant properties: the first is attributed to its chemical structure, which enables it to directly neutralize reactive oxygen species (ROS); the second is related to its ability to induce the expression of antioxidant enzymes via the transcription factor nuclear factor erythroid 2–related factor 2 (Nrf2). Both ROS and Nrf2 are closely associated with mitochondrial function and metabolism, and their dysregulation may lead to mitochondrial dysfunction, potentially contributing to the development of various pathological conditions. Therefore, curcumin treatment appears highly promising and is strongly associated with the preservation of mitochondrial function. The aim of this review is to summarize the current literature on the impact of curcumin’s antioxidant properties on mitochondrial function. Specifically, studies conducted in different biological models are included, with emphasis on aspects such as mitochondrial respiration, antioxidant enzyme activity, interactions with mitochondrial membranes, and the role of curcumin in the regulation of intrinsic apoptosis. Full article
Show Figures

Figure 1

27 pages, 12231 KB  
Review
Mitochondria-Associated Membrane Dysfunction in Neurodegeneration and Its Effects on Lipid Metabolism, Calcium Signaling, and Cell Fate
by Thi Thuy Truong, Alka Ashok Singh, Nguyen Van Bang, Nguyen Minh Hung Vu, Sungsoo Na, Jaeyeop Choi, Junghwan Oh and Sudip Mondal
Membranes 2025, 15(9), 263; https://doi.org/10.3390/membranes15090263 (registering DOI) - 31 Aug 2025
Abstract
Mitochondria-associated membranes (MAMs) are essential for cellular homeostasis. MAMs are specialized contact sites located between the endoplasmic reticulum (ER) and mitochondria and control apoptotic pathways, lipid metabolism, autophagy initiation, and calcium signaling, processes critical to the survival and function of neurons. Although this [...] Read more.
Mitochondria-associated membranes (MAMs) are essential for cellular homeostasis. MAMs are specialized contact sites located between the endoplasmic reticulum (ER) and mitochondria and control apoptotic pathways, lipid metabolism, autophagy initiation, and calcium signaling, processes critical to the survival and function of neurons. Although this area of membrane biology remains understudied, increasing evidence links MAM dysfunction to the etiology of major neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis (ALS). MAMs consist of a network of protein complexes that mediate molecular exchange and ER–mitochondria tethering. MAMs regulate lipid flow in the brain, including phosphatidylserine and cholesterol; disruption of this process causes membrane instability and impaired synaptic function. Inositol 1,4,5-trisphosphate receptor—voltage-dependent anion channel 1 (IP3R-VDAC1) interactions at MAMs maintain calcium homeostasis, which is required for mitochondria to produce ATP; dysregulation promotes oxidative stress and neuronal death. An effective therapeutic approach for altering neurodegenerative processes is to restore the functional integrity of MAMs. Improving cell-to-cell interactions and modulating MAM-associated proteins may contribute to the restoration of calcium homeostasis and lipid metabolism, both of which are key for neuronal protection. MAMs significantly contribute to the progression of neurodegenerative diseases, making them promising targets for future therapeutic research. This review emphasizes the increasing importance of MAMs in the study of neurodegeneration and their potential as novel targets for membrane-based therapeutic interventions. Full article
(This article belongs to the Section Biological Membranes)
Show Figures

Figure 1

34 pages, 2865 KB  
Review
Mitochondrial Transport Proteins in Cardiovascular Diseases: Metabolic Gatekeepers, Pathogenic Mediators and Therapeutic Targets
by Yue Pei, Sitong Wan, Jingyi Qi, Xueyao Xi, Yinhua Zhu, Peng An, Junjie Luo and Yongting Luo
Int. J. Mol. Sci. 2025, 26(17), 8475; https://doi.org/10.3390/ijms26178475 (registering DOI) - 31 Aug 2025
Abstract
Mitochondria, as the metabolic hubs of cells, play a pivotal role in maintaining cardiovascular homeostasis through dynamic regulation of energy metabolism, redox balance, and calcium signaling. Cardiovascular diseases (CVDs), including heart failure, ischemic heart disease, cardiomyopathies, and myocardial infarction, remain the leading cause [...] Read more.
Mitochondria, as the metabolic hubs of cells, play a pivotal role in maintaining cardiovascular homeostasis through dynamic regulation of energy metabolism, redox balance, and calcium signaling. Cardiovascular diseases (CVDs), including heart failure, ischemic heart disease, cardiomyopathies, and myocardial infarction, remain the leading cause of global mortality, with mitochondrial dysfunction emerging as a unifying pathological mechanism across these conditions. Emerging evidence suggests that impaired mitochondrial transport systems—critical gatekeepers of metabolite flux, ion exchange, and organelle communication—drive disease progression by disrupting bioenergetic efficiency and exacerbating oxidative stress. This review synthesizes current knowledge on mitochondrial transport proteins, such as the voltage-dependent anion channels, transient receptor potential channels, mitochondrial calcium uniporter, and adenine nucleotide translocator, focusing on their structural–functional relationships and dysregulation in CVD pathogenesis. We highlight how aberrant activity of these transporters contributes to hallmark features of cardiac pathology, including metabolic inflexibility, mitochondrial permeability transition pore destabilization, and programmed cell death. Furthermore, we critically evaluate preclinical advances in targeting mitochondrial transport systems through pharmacological modulation, gene editing, and nanoparticle-based delivery strategies. By elucidating the mechanistic interplay between transport protein dysfunction and cardiac metabolic reprogramming, we address a critical knowledge gap in cardiovascular biology and provide a roadmap for developing precision therapies. Our insights underscore the translational potential of mitochondrial transport machinery as both diagnostic biomarkers and therapeutic targets, offering new avenues to combat the growing burden of CVDs in aging populations. Full article
(This article belongs to the Special Issue Mitochondria in Aging and Aging-Related Diseases)
Show Figures

Figure 1

26 pages, 1536 KB  
Review
Generalized Anxiety Disorder and Obesity: Overlapping Neuroendocrine, Metabolic, and Behavioral Pathways
by Agnieszka Dymek, Magdalena Zielińska, Anna Englert-Bator, Katarzyna Dereń and Edyta Łuszczki
Nutrients 2025, 17(17), 2835; https://doi.org/10.3390/nu17172835 (registering DOI) - 31 Aug 2025
Abstract
Background/Objectives: Generalized anxiety disorder (GAD) is one of the most commonly diagnosed anxiety disorders in primary care. The global lifetime prevalence of GAD is estimated at 3.7%, ranging from 1.6% in low-income countries to 5.0% in high-income countries, underscoring its widespread impact. [...] Read more.
Background/Objectives: Generalized anxiety disorder (GAD) is one of the most commonly diagnosed anxiety disorders in primary care. The global lifetime prevalence of GAD is estimated at 3.7%, ranging from 1.6% in low-income countries to 5.0% in high-income countries, underscoring its widespread impact. Given the frequent co-occurrence of GAD with obesity, this association has important clinical implications, particularly for screening, prevention, and treatment strategies. The aim of this review is to identify potential biological mechanisms linking obesity and GAD, summarize the current state of knowledge in this area, and highlight existing research gaps, as well as directions for future research. Methods: This narrative review is based on the literature published between 2015 and 2025 concerning the co-occurrence of GAD and obesity, with a focus on potential shared mechanisms including HPA axis dysregulation, chronic inflammation, oxidative stress, insulin resistance, gut–brain axis and microbiota dysbiosis, sleep disturbance, and maladaptive eating behaviors. Results/Conclusions: A growing body of evidence suggests an important, albeit still ambiguously defined, relationship between obesity and GAD. GAD and obesity may reinforce each other, leading to a mutually reinforcing relationship. Despite growing interest, high-quality prospective and interventional studies focusing specifically on GAD are lacking. A potentially effective therapeutic approach should be integrated and multidisciplinary, combining psychological, pharmacological, and lifestyle interventions. It may also be beneficial for clinicians to consider routine assessment of anxiety in patients with obesity and, conversely, to monitor metabolic risk in individuals with GAD. Such an approach, targeting both mental and metabolic domains, holds promise for improving outcomes. Full article
Show Figures

Figure 1

23 pages, 1099 KB  
Article
From Systemic Inflammation to Vascular Remodeling: Investigating Carotid IMT in COVID-19 Survivors
by Emilia Bielecka, Piotr Sielatycki, Paulina Pietraszko, Sara Anna Frankowska and Edyta Zbroch
Viruses 2025, 17(9), 1196; https://doi.org/10.3390/v17091196 (registering DOI) - 30 Aug 2025
Abstract
Background: Atherosclerosis is a chronic inflammatory condition that underlies both cardiovascular and cerebrovascular complications. Emerging evidence suggests that COVID-19 may play a role in its progression. Purpose: The aim of the study was to evaluate the potential impact of SARS-CoV-2 infection on the [...] Read more.
Background: Atherosclerosis is a chronic inflammatory condition that underlies both cardiovascular and cerebrovascular complications. Emerging evidence suggests that COVID-19 may play a role in its progression. Purpose: The aim of the study was to evaluate the potential impact of SARS-CoV-2 infection on the development of atherosclerosis. Patients and Methods: Common carotid artery (CCA) intima media thickness (IMT) was measured by ultrasonography twice, 12–18 months apart, in a cohort of 92 patients (47 with COVID-19 and 45 controls). Clinical data were collected from medical histories, physical examinations, and laboratory findings. Results: Baseline IMT values were comparable between the study groups (0.85 mm vs. 0.78 mm). However, the COVID-19 group exhibited a significantly greater increase in IMT over time, with a median change of 0.13 mm compared to 0.05 mm in the controls (p = 0.018). Furthermore, 69.2% of COVID-19 patients exceeded the median IMT progression threshold compared to 36% in the control group (p = 0.017). An elevated level of C-reactive protein (CRP) and a higher triglyceride (Tg)-to-High-Density Lipoprotein Cholesterol (HDL) ratio were significantly associated with increased IMT in the COVID-19 group. Age and heart rate were identified as significant predictors of IMT progression across both groups. Conclusion: COVID-19 may accelerate the progression of subclinical atherosclerosis. The strong associations of CRP and the TG/HDL ratio with IMT highlight the potential roles of chronic inflammation and metabolic dysregulation in driving these vascular changes. Further large-scale, multicenter studies are needed to elucidate the underlying mechanisms, confirm these observations, and guide targeted preventive and therapeutic strategies for individuals with an increased cardiovascular and cerebrovascular risk. Full article
(This article belongs to the Special Issue COVID-19 Complications and Co-infections)
24 pages, 2159 KB  
Article
Agentic RAG-Driven Multi-Omics Analysis for PI3K/AKT Pathway Deregulation in Precision Medicine
by Micheal Olaolu Arowolo, Sulaiman Olaniyi Abdulsalam, Rafiu Mope Isiaka, Kingsley Theophilus Igulu, Bukola Fatimah Balogun, Mihail Popescu and Dong Xu
Algorithms 2025, 18(9), 545; https://doi.org/10.3390/a18090545 (registering DOI) - 30 Aug 2025
Abstract
The phosphoinositide 3-kinase (PI3K)/AKT signaling pathway is a crucial regulator of cellular metabolism, proliferation, and survival. It is frequently dysregulated in metabolic, cardiovascular, and neoplastic disorders. Despite the advancements in multi-omics technology, existing methods often fail to provide real-time, pathway-specific insights for precision [...] Read more.
The phosphoinositide 3-kinase (PI3K)/AKT signaling pathway is a crucial regulator of cellular metabolism, proliferation, and survival. It is frequently dysregulated in metabolic, cardiovascular, and neoplastic disorders. Despite the advancements in multi-omics technology, existing methods often fail to provide real-time, pathway-specific insights for precision medicine and drug repurposing. We offer Agentic RAG-Driven Multi-Omics Analysis (ARMOA), an autonomous, hypothesis-driven system that integrates retrieval-augmented generation (RAG), large language models (LLMs), and agentic AI to thoroughly analyze genomic, transcriptomic, proteomic, and metabolomic data. Through the use of graph neural networks (GNNs) to model complex interactions within the PI3K/AKT pathway, ARMOA enables the discovery of novel biomarkers, probable candidates for drug repurposing, and customized therapy responses to address the complexities of PI3K/AKT dysregulation in disease states. ARMOA dynamically gathers and synthesizes knowledge from multiple sources, including KEGG, TCGA, and DrugBank, to guarantee context-aware insights. Through adaptive reasoning, it gradually enhances predictions, achieving 91% accuracy in external testing and 92% accuracy in cross-validation. Case studies in breast cancer and type 2 diabetes demonstrate that ARMOA can identify synergistic drug combinations with high clinical relevance and predict therapeutic outcomes specific to each patient. The framework’s interpretability and scalability are greatly enhanced by its use of multi-omics data fusion and real-time hypothesis creation. ARMOA provides a cutting-edge example for precision medicine by integrating multi-omics data, clinical judgment, and AI agents. Its ability to provide valuable insights on its own makes it a powerful tool for advancing biomedical research and treatment development. Full article
(This article belongs to the Special Issue Advanced Algorithms for Biomedical Data Analysis)
Show Figures

Figure 1

41 pages, 2467 KB  
Review
Crosstalk Between Skeletal Muscle and Proximal Connective Tissues in Lipid Dysregulation in Obesity and Type 2 Diabetes
by Nataša Pollak, Efua Gyakye Janežič, Žiga Šink and Chiedozie Kenneth Ugwoke
Metabolites 2025, 15(9), 581; https://doi.org/10.3390/metabo15090581 (registering DOI) - 30 Aug 2025
Abstract
Background/Objectives: Obesity and type 2 diabetes mellitus (T2DM) profoundly disrupt lipid metabolism within local microenvironments of skeletal muscle and its associated connective tissues, including adipose tissue, bone, and fascia. However, the role of local communication between skeletal muscle and its proximal connective tissues [...] Read more.
Background/Objectives: Obesity and type 2 diabetes mellitus (T2DM) profoundly disrupt lipid metabolism within local microenvironments of skeletal muscle and its associated connective tissues, including adipose tissue, bone, and fascia. However, the role of local communication between skeletal muscle and its proximal connective tissues in propagating metabolic dysfunction is incompletely understood. This narrative review synthesizes current evidence on these local metabolic interactions, highlighting novel insights and existing gaps. Methods: We conducted a comprehensive literature analysis of primary research published in the last decade, sourced from PubMed, Web of Science, and ScienceDirect. Studies were selected for relevance to skeletal muscle, adipose tissue, fascia, and bone lipid metabolism in the context of obesity and T2DM, with emphasis on molecular, cellular, and paracrine mechanisms of local crosstalk. Findings were organized into thematic sections addressing physiological regulation, pathological remodeling, and inter-organ signaling pathways. Results: Our synthesis reveals that local lipid dysregulation in obesity and T2DM involves altered fatty acid transporter dynamics, mitochondrial overload, fibro-adipogenic remodeling, and compartment-specific adipose tissue dysfunction. Crosstalk via myokines, adipokines, osteokines, bioactive lipids, and exosomal miRNAs integrates metabolic responses across these tissues, amplifying insulin resistance and lipotoxic stress. Emerging evidence highlights the underappreciated roles of fascia and marrow adipocytes in regional lipid handling. Conclusions: Collectively, these insights underscore the pivotal role of inter-tissue crosstalk among skeletal muscle, adipose tissue, bone, and fascia in orchestrating lipid-induced insulin resistance, and highlight the need for integrative strategies that target this multicompartmental network to mitigate metabolic dysfunction in obesity and T2DM. Full article
(This article belongs to the Special Issue Lipid Metabolism Disorders in Obesity)
Show Figures

Figure 1

18 pages, 2636 KB  
Article
Urine Metabolomics of Gout Reveals the Dynamic Reprogramming and Non-Invasive Biomarkers of Disease Progression
by Guizhen Zhu, Yuan Luo, Nan Su, Xiangyi Zheng, Zhusong Mei, Qiao Ye, Jie Peng, Peiyu An, Yangqian Song, Weina Luo, Hongxia Li, Guangyun Wang and Haitao Zhang
Metabolites 2025, 15(9), 580; https://doi.org/10.3390/metabo15090580 - 29 Aug 2025
Abstract
Background/Objectives: Gout, a complex metabolic disorder of increasing global incidence, remains incompletely understood in its pathogenesis. Current diagnostic approaches exhibit significant limitations, including insufficient specificity and the requirement for invasive joint aspiration, highlighting the need for non-invasive, sensitive biomarkers for early detection. Methods: [...] Read more.
Background/Objectives: Gout, a complex metabolic disorder of increasing global incidence, remains incompletely understood in its pathogenesis. Current diagnostic approaches exhibit significant limitations, including insufficient specificity and the requirement for invasive joint aspiration, highlighting the need for non-invasive, sensitive biomarkers for early detection. Methods: Urine metabolites were extracted from 28 healthy controls, 13 asymptomatic hyperuricemia (HUA) patients, and 29 acute gouty arthritis (AGA) patients. The extracted metabolites were analyzed by UHPLC-MS/MS for untargeted metabolomics. Differential metabolites were screened by partial least squares discriminant analysis (PLS-DA) and volcano plot analysis. Pathway analysis determined the core disorder pathway of gout progression. Results: A total of 278 differential metabolites associated with gout progression were identified. The most pronounced metabolic alterations were observed between the AGA and control groups, indicative of substantial metabolic reprogramming during disease transition. Metabolic pathway analysis revealed four significantly dysregulated pathways: histidine metabolism, nicotinate and nicotinamide metabolism, phenylalanine metabolism, and tyrosine metabolism. Receiver operating characteristic (ROC) curve analysis revealed that three urine markers with high diagnostic efficacy—oxoamide, 3-methylindole, and palmitic acid—exhibited progressive alterations across the disease continuum. Conclusions: This metabolomics study identified core regulatory metabolites and newly discovered metabolic pathways underlying gout pathogenesis, along with novel urinary biomarkers capable of predicting HUA-to-AGA progression. The aberrant levels of key metabolites in the disordered pathway implicate neuroimmune dysregulation, energy metabolism disruption, and oxidative stress in gout pathogenesis. These findings provide new foundations and strategies for the daily monitoring and prevention of gout. Full article
22 pages, 1862 KB  
Article
Effects of Dietary and Probiotic Interventions in Patients with Metabolic Syndrome and Obstructive Sleep Apnea
by Amina Venter, Amin-Florin El-kharoubi, Mousa El-kharoubi, Evelin Claudia Ghitea, Marc Cristian Ghitea, Timea Claudia Ghitea and Ciprian Florian Venter
Clin. Pract. 2025, 15(9), 159; https://doi.org/10.3390/clinpract15090159 - 29 Aug 2025
Abstract
Background: Metabolic syndrome (MS) and obstructive sleep apnea (OSA) frequently coexist, exacerbating systemic inflammation, oxidative stress, and metabolic dysregulation. This study evaluates the effects of dietary and probiotic interventions, compared to a non-intervention control group, on metabolic, hemodynamic, and neurochemical parameters, with a [...] Read more.
Background: Metabolic syndrome (MS) and obstructive sleep apnea (OSA) frequently coexist, exacerbating systemic inflammation, oxidative stress, and metabolic dysregulation. This study evaluates the effects of dietary and probiotic interventions, compared to a non-intervention control group, on metabolic, hemodynamic, and neurochemical parameters, with a specific focus on the neurotransmitters GABA and glutamate. Methods: In a prospective randomized study (2020–2023), 120 patients with coexisting MS and OSA were assigned to three groups: control (n = 36), diet therapy (n = 42), and diet therapy combined with probiotics (n = 42). Interventions lasted six months and included personalized dietary plans and probiotic supplementation. Outcome measures included BMI, visceral fat, HOMA index, lipid profile, oxygen saturation, and urinary GABA and glutamate levels. Unsupervised K-means clustering and principal component analysis (PCA) were applied to identify phenotypic response patterns based on delta values. Results: Diet therapy led to significant reductions in BMI (−15.7%, p = 0.001), visceral fat (−17.3%, p = 0.001), triglycerides (−14.6%, p = 0.003), uric acid (−9.5%, p = 0.011), and C-reactive protein (CRP) (−21.4%, p = 0.007). The combined intervention group exhibited further improvements in visceral fat (−22.8%, p = 0.001), glutamate (−18.2%, p = 0.002), and GABA levels (+19.5%, p = 0.001). Oxygen saturation improved across all groups, with the greatest increase in the probiotics group (+2.3%). Clustering analysis revealed three distinct response phenotypes—strong, moderate, and non-responders—highlighting inter-individual variability in treatment efficacy. Conclusions: Personalized dietary interventions, especially when paired with probiotics, effectively improve metabolic, inflammatory, and neurochemical profiles in patients with MS and OSA. Integrating clustering algorithms enables phenotype-specific stratification, offering a step toward precision lifestyle medicine. Future studies should explore long-term outcomes and refine microbiota-targeted approaches to optimize intervention efficacy. Full article
(This article belongs to the Special Issue The Effect of Dietary Compounds on Inflammation-Mediated Diseases)
25 pages, 1023 KB  
Review
Beyond Support Cells: Astrocytic Autophagy as a Central Regulator of CNS Homeostasis and Neurodegenerative Diseases
by Jung Ho Lee, Wonseok Chang, Sun Seek Min, Dae Yong Song and Hong Il Yoo
Cells 2025, 14(17), 1342; https://doi.org/10.3390/cells14171342 - 29 Aug 2025
Abstract
Autophagy is a fundamental catabolic pathway critical for maintaining cellular homeostasis in the central nervous system (CNS). While neuronal autophagy has been extensively studied, growing evidence highlights the crucial roles of astrocytic autophagy in CNS physiology and pathology. Astrocytes regulate metabolic support, redox [...] Read more.
Autophagy is a fundamental catabolic pathway critical for maintaining cellular homeostasis in the central nervous system (CNS). While neuronal autophagy has been extensively studied, growing evidence highlights the crucial roles of astrocytic autophagy in CNS physiology and pathology. Astrocytes regulate metabolic support, redox balance, and neuroinflammatory responses. These functions are closely linked to autophagic activity. The disruption of astrocytic autophagy contributes to synaptic dysfunction, chronic inflammation, myelin impairment, and blood–brain barrier instability. Dysregulation of astrocytic autophagy has been implicated in the pathogenesis of multiple neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. This review summarizes the molecular mechanisms of autophagy in astrocytes and delineates its role in intercellular communication with neurons, microglia, oligodendrocytes, and endothelial cells. Furthermore, we will discuss current pharmacological approaches targeting astrocytic autophagy, with particular attention to repurposed agents such as rapamycin, lithium, and caloric restriction mimetics. Although promising in preclinical models, therapeutic translation is challenged by the complexity of autophagy’s dual roles and cell-type specificity. A deeper understanding of astrocytic autophagy and its crosstalk with other CNS cell types may facilitate the development of targeted interventions for neurodegenerative diseases. Full article
(This article belongs to the Special Issue The Role Glial Cells in Neurodegenerative Disorders)
19 pages, 570 KB  
Review
Imaging of Cerebral Iron as an Emerging Marker for Brain Aging, Neurodegeneration, and Cerebrovascular Diseases
by Chi-Heng Zhou and Yi-Cheng Zhu
Brain Sci. 2025, 15(9), 944; https://doi.org/10.3390/brainsci15090944 - 29 Aug 2025
Abstract
Iron is critical for brain development, metabolism, and function; however, dysregulated iron disposition contributes to neurological diseases. Many neuroimaging techniques have enabled detection of iron susceptibility, and quantitative susceptibility mapping (QSM) offers a sensitive magnetic resonance imaging (MRI) technique for quantifying brain iron. [...] Read more.
Iron is critical for brain development, metabolism, and function; however, dysregulated iron disposition contributes to neurological diseases. Many neuroimaging techniques have enabled detection of iron susceptibility, and quantitative susceptibility mapping (QSM) offers a sensitive magnetic resonance imaging (MRI) technique for quantifying brain iron. To elucidate the functional role of cerebral iron and clarify the utility of QSM in establishing iron as a potential biomarker, this review synthesizes cellular and regional behaviours of iron from physiological aging to disease conditions, with a focus on neurodegeneration such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and multiple sclerosis (MS), as well as cerebral small vessel disease (CSVD) as cerebrovascular manifestation. Distinct patterns of iron distribution in deep gray matter and selective cortical regions are associated with motor and cognitive impairment, while the interaction between iron, vascular integrity, and glial function further stresses its pathological relevance. QSM of iron may, thereby, serve as a marker to monitor iron-related disease progression and facilitate intervention. Temporal dynamics of iron in brain pathology remain underexplored, and we emphasized the need for longitudinal mapping and multi-modality biomarker integration. Establishing iron as a clinically relevant imaging biomarker requires continued investigation into its topographical, molecular, and functional correlates across aging and disease trajectories. Full article
(This article belongs to the Special Issue Using Neuroimaging to Explore Neurodegenerative Diseases)
Show Figures

Figure 1

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 68
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

30 pages, 1131 KB  
Review
Extra-Ribosomal Roles for Ribosomal Proteins and Their Relevance to Tumour Suppression, Carcinogenesis and Cancer Progression
by Sreepriya Pk, Joshua Freimanis, Thomas Kovala and Amadeo M. Parissenti
Cancers 2025, 17(17), 2825; https://doi.org/10.3390/cancers17172825 - 29 Aug 2025
Viewed by 62
Abstract
Protein translation by ribosomes is one of the most energetically costly cellular processes. Consequently, the number and activity of ribosomes in cells and tissues are precisely tailored to match metabolic demands. While ribosomal proteins (RPs) play essential roles in facilitating and regulating the [...] Read more.
Protein translation by ribosomes is one of the most energetically costly cellular processes. Consequently, the number and activity of ribosomes in cells and tissues are precisely tailored to match metabolic demands. While ribosomal proteins (RPs) play essential roles in facilitating and regulating the translation of mRNA transcripts into protein, there is increasing evidence that free RPs not bound to ribosomes can play important roles in cellular regulation. Often, free RPs act as tumour suppressors by multiple mechanisms, for example, by inducing cell cycle arrest through their ability to bind and inhibit MDM2-mediated p53 degradation. Dysregulation of these RPs, however, can result in various diseases like Diamond-Blackfan anemia, ribosomopathies, and other diseases. In cancer, epigenetic modifications, altered transcription, and processing defects in the rRNAs create “onco-ribosomes” that strongly support tumour cell replication, invasion and metastasis. In this context, free RPs in tumour cells (often mutated or post-translationally modified) further promote tumour cell proliferation, invasion, and metastasis. This review focuses specifically on extra-ribosomal roles for RPs, where depending upon cellular context, they act outside of the ribosome to either suppress tumorigenesis in normal tissues or promote tumour proliferation and progression. This new understanding of the interplay between RPs and pathways suppressing or promoting tumorigenesis further emphasizes why the ribosome is increasingly being seen as an important therapeutic target in human cancers. Full article
Show Figures

Figure 1

17 pages, 3785 KB  
Article
Peroxisome Proliferator-Activated Receptor Family of Lipid-Activated Nuclear Receptors Alpha Silencing Promotes Oxidative Stress and Hypertrophic Phenotype in Rat Cardiac Cells
by Marzia Bianchi, Nadia Panera, Sara Petrillo, Nicolò Cicolani, Cristiano De Stefanis, Marco Scarsella, Domenico Ciavardelli, Fiorella Piemonte, Anna Alisi and Anna Pastore
Antioxidants 2025, 14(9), 1059; https://doi.org/10.3390/antiox14091059 - 28 Aug 2025
Viewed by 74
Abstract
The peroxisome proliferator-activated receptor family of lipid-activated nuclear receptors (PPARs) plays a critical role in the regulation of cellular lipid metabolism. In cardiac muscle, PPARα is highly expressed and regulates genes involved in fatty acid oxidation, but its activity is downregulated in hypertrophic [...] Read more.
The peroxisome proliferator-activated receptor family of lipid-activated nuclear receptors (PPARs) plays a critical role in the regulation of cellular lipid metabolism. In cardiac muscle, PPARα is highly expressed and regulates genes involved in fatty acid oxidation, but its activity is downregulated in hypertrophic hearts; however, the consequences of chronic PPARα deficiency on the cardiac contractile apparatus remain unclear. This study aimed to investigate the PPARα role in hypertrophic phenotype and to evaluate the potential effects of the antioxidant Ebselen (Ebs) treatment on changes associated with PPARα depletion. We thus generated an in vitro model of cardiac hypertrophy by stable silencing of the PPARA gene in H9c2 rat cardiomyoblasts. We observed that PPARα silencing induces a hypertrophic phenotype, characterized by increased NPPB and decreased FBXO32 expression, mitochondrial dysregulation, impaired lipid metabolism, oxidative stress, and ferroptosis-related alterations. Epigenetically, H3K27ac levels increased while H3K27me3 decreased. Moreover, miR-34a, miR-132, and miR-331 were downregulated, implicating a miRNA-mediated mechanism in PPARα-linked cardiac hypertrophy. Treatment with Ebs, a redox-active compound with inhibitory effects on ferroptosis and epigenetics, reversed hypertrophic phenotype and restored miRNA levels. In conclusion, we found that PPARα depletion promotes oxidative stress and hypertrophic phenotype and that Ebs may act as a potential therapeutic agent. Full article
Show Figures

Figure 1

20 pages, 936 KB  
Review
The Mediator Complex: A Regulatory Hub for Transcriptional Activity of Nuclear Receptors
by Liming Zhou, Manhan Zhao, Yifei Zhai and Qiong Lin
Cells 2025, 14(17), 1335; https://doi.org/10.3390/cells14171335 - 28 Aug 2025
Viewed by 202
Abstract
The Mediator complex plays a key role in gene transcription. In particular, the interaction of the Mediator complex with nuclear receptors, the known transcription factors, regulates multiple nuclear receptor-mediated gene transcription pathways and associated cellular functions. Dysregulation of the interaction of the Mediator [...] Read more.
The Mediator complex plays a key role in gene transcription. In particular, the interaction of the Mediator complex with nuclear receptors, the known transcription factors, regulates multiple nuclear receptor-mediated gene transcription pathways and associated cellular functions. Dysregulation of the interaction of the Mediator complex with nuclear receptors results in many pathological processes, such as cancer, metabolic and neuronal diseases. Thus, understanding of the mechanism by which the Mediator complex regulates the nuclear receptor-mediated transcriptional activity and biological function is crucial for therapy of both the Mediator complex- and nuclear receptor-associated diseases. In this review article, we attempt to summarize current research progress in the interaction of the Mediator complex with nuclear receptors and the associated nuclear receptor transcriptional signaling pathways, explore the clinical potential of the Mediator complex as a therapeutic target, and provide new perspectives for the treatment of diseases associated with the Mediator complex and nuclear receptors. Full article
Show Figures

Figure 1

Back to TopTop