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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (186)

Search Parameters:
Keywords = non-erythroid

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 3599 KB  
Article
Antioxidant Intervention in NAFLD: Astaxanthin and Kokum Modulate Redox Status and Lysosomal Degradation
by Natalia Ksepka, Natalia Kuzia, Sara Frazzini, Luciana Rossi, Małgorzata Łysek-Gładysińska, Michał Ławiński and Artur Jóźwik
Molecules 2026, 31(2), 321; https://doi.org/10.3390/molecules31020321 - 16 Jan 2026
Viewed by 120
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major metabolic disorder characterized by hepatic lipid accumulation, oxidative stress, and disturbance of lysosomal degradation. Central to these processes is glutathione (GSH), a key antioxidant regulating redox balance and cellular homeostasis. This study aimed to evaluate [...] Read more.
Non-alcoholic fatty liver disease (NAFLD) is a major metabolic disorder characterized by hepatic lipid accumulation, oxidative stress, and disturbance of lysosomal degradation. Central to these processes is glutathione (GSH), a key antioxidant regulating redox balance and cellular homeostasis. This study aimed to evaluate the therapeutic potential of two dietary antioxidants—astaxanthin and Garcinia indica (kokum)—in modulating hepatic redox status, lysosomal function, and metabolic gene expression in a murine model of diet-induced NAFLD. A total of 120 male Swiss Webster mice were allocated into control and steatotic groups, followed by a 90-day supplementation period with astaxanthin, kokum, or their combination. Liver tissue was collected post-supplementation for biochemical, antioxidant, and qRT-PCR analyses. Outcomes included lysosomal enzymes activities, superoxide dismutase (SOD), GSH, vitamin C, total polyphenols, DPPH radical-scavenging activity, and total antioxidant capacity (TAC). NAFLD induced marked oxidative stress, lysosomal overactivation, and alteration of antioxidant-related gene expression. Combined supplementation restored GSH, enhanced TAC, reduced lysosomal stress markers, and significantly upregulated nuclear factor erythroid 2-related factor 2 (Nfe2l2) while downregulating fatty acid synthase (FASN) and partially rescuing lipoprotein lipase (LpL). Correlation analyses revealed strong associations between antioxidant capacity, lysosomal function, and transcriptional regulation, supporting the therapeutic relevance of combined antioxidant therapy for concurrent redox and lysosomal dysregulation in NAFLD. These findings underscore the therapeutic potential of targeting redox and cellular degradation pathways with antioxidant-based interventions to re-establish hepatic metabolic balance in NAFLD and related disorders. Full article
(This article belongs to the Special Issue Antioxidant, and Anti-Inflammatory Activities of Natural Plants)
Show Figures

Graphical abstract

23 pages, 2788 KB  
Article
Molecular Insights into the Synergistic Anticancer and Oxidative Stress–Modulating Activity of Quercetin and Gemcitabine
by Yasemin Afşin, Senem Alkan Akalın, İlhan Özdemir, Mehmet Cudi Tuncer and Şamil Öztürk
Antioxidants 2026, 15(1), 91; https://doi.org/10.3390/antiox15010091 - 10 Jan 2026
Viewed by 307
Abstract
Quercetin (Q), a bioactive flavonoid, exerts potent antioxidant and redox-modulating effects by activating the nuclear factor erythroid 2-related factor 2/antioxidant response Element (Nrf2/ARE) pathway and upregulating endogenous antioxidant defenses, including enzymatic antioxidants such as superoxide dismutase (SOD) and catalase (CAT), as well as [...] Read more.
Quercetin (Q), a bioactive flavonoid, exerts potent antioxidant and redox-modulating effects by activating the nuclear factor erythroid 2-related factor 2/antioxidant response Element (Nrf2/ARE) pathway and upregulating endogenous antioxidant defenses, including enzymatic antioxidants such as superoxide dismutase (SOD) and catalase (CAT), as well as non-enzymatic glutathione (GSH) and lipid peroxidation (MDA). Gemcitabine (Gem), a widely used antimetabolite chemotherapeutic, often shows limited efficacy under hypoxic and oxidative stress conditions driven by hypoxia-inducible factor 1-alpha (HIF-1α) and vascular endothelial growth factor (VEGF)-mediated angiogenesis. This study investigated the redox-mediated synergistic effects of Q and Gem in MDA-MB-231 human breast cancer cells. Combination treatment significantly reduced cell viability beyond the expected Bliss value, indicating a synergistic interaction and enhanced apoptosis compared with single-agent treatments. Increased reactive oxygen species (ROS) production was accompanied by depletion of GSH and accumulation of MDA, establishing a pro-apoptotic oxidative stress environment. Q alone enhanced SOD and CAT activities, whereas the combination induced exhaustion of antioxidant defenses under oxidative load, reflecting a redox-adaptive response. Molecular analyses revealed downregulation of HIF-1α and VEGF, alongside upregulation of Bax and Caspase-3, confirming suppression of hypoxia-driven survival and activation of the intrinsic apoptotic pathway. Transcriptomic and enrichment analyses further identified modulation of oxidative stress- and apoptosis-related pathways, including phosphoinositide-3-kinase–protein kinase B/Akt (PI3K/Akt), HIF-1 and VEGF signaling. Collectively, these results indicate that Q potentiates Gem cytotoxicity via redox modulation, promoting controlled ROS elevation and apoptosis while suppressing hypoxia-induced survival mechanisms, highlighting the therapeutic potential of redox-based combination strategies against chemoresistant breast cancer. Full article
(This article belongs to the Special Issue Redox Biomarkers in Cancer)
Show Figures

Figure 1

30 pages, 2720 KB  
Review
Nutritional Regulation of Cardiac Metabolism and Function: Molecular and Epigenetic Mechanisms and Their Role in Cardiovascular Disease Prevention
by Lucia Capasso, Donato Mele, Rosaria Casalino, Gregorio Favale, Giulia Rollo, Giulia Verrilli, Mariarosaria Conte, Paola Bontempo, Vincenzo Carafa, Lucia Altucci and Angela Nebbioso
Nutrients 2026, 18(1), 93; https://doi.org/10.3390/nu18010093 - 27 Dec 2025
Viewed by 582
Abstract
Background: Cardiovascular diseases (CVDs) remain the leading cause of mortality worldwide and are strongly influenced by dietary habits. Beyond caloric intake, nutrients act as molecular signals that regulate cardiac metabolism, mitochondrial function, inflammation, and epigenetic remodeling. Objectives: This review aims to synthesize [...] Read more.
Background: Cardiovascular diseases (CVDs) remain the leading cause of mortality worldwide and are strongly influenced by dietary habits. Beyond caloric intake, nutrients act as molecular signals that regulate cardiac metabolism, mitochondrial function, inflammation, and epigenetic remodeling. Objectives: This review aims to synthesize current evidence on how dietary patterns and specific nutritional interventions regulate cardiac metabolism and function through interconnected molecular and epigenetic mechanisms, highlighting their relevance for cardiovascular disease prevention. Methods: A narrative review of the literature was conducted using PubMed, Scopus, and Web of Science, focusing on studies published between 2006 and 2025. Experimental, translational, and clinical studies addressing diet-induced modulation of cardiac metabolic pathways, oxidative and inflammatory signaling, epigenetic regulation, and gut microbiota-derived metabolites were included. Results: The analyzed literature consistently shows that unbalanced diets rich in saturated fats and refined carbohydrates impair cardiac metabolic flexibility by disrupting key nutrient-sensing pathways, including AMP-activated protein kinase (AMPK), proliferator-activated receptor alpha (PPARα), mammalian target of rapamycin (mTOR), and sirtuin 1/peroxisome proliferator-activated receptor gamma coactivator 1-alpha (SIRT1/PGC-1α), leading to mitochondrial dysfunction, oxidative stress, chronic inflammation, and maladaptive remodeling. In contrast, cardioprotective dietary patterns, such as caloric restriction (CR), intermittent fasting (IF), and Mediterranean and plant-based diets, enhance mitochondrial efficiency, redox balance, and metabolic adaptability. These effects are mediated by coordinated activation of AMPK-SIRT1 signaling, suppression of mTOR over-activation, modulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways, and favorable epigenetic remodeling involving DNA methylation, histone modifications, and non-coding RNAs. Emerging evidence also highlights the central role of gut microbiota-derived metabolites, particularly short-chain fatty acids, in linking diet to epigenetic and metabolic regulation of cardiac function. Conclusions: Diet quality emerges as a key determinant of cardiac metabolic health, acting through integrated molecular, epigenetic, and microbiota-mediated mechanisms. Targeted nutritional strategies can induce long-lasting cardioprotective metabolic and epigenetic adaptations, supporting the concept of diet as a modifiable molecular intervention. These findings provide a mechanistic rationale for integrating personalized nutrition into cardiovascular prevention and precision cardiology, complementing standard pharmacological therapies. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Diet-Associated Cardiac Metabolism)
Show Figures

Figure 1

28 pages, 19507 KB  
Article
Parvovirus B19 and Cellular Transcriptome Dynamics in Differentiating Erythroid Progenitor Cells
by Erika Fasano, Niccolò Guglietta, Federica Bichicchi, Ilaria Gasperini, Elisabetta Manaresi and Giorgio Gallinella
Viruses 2026, 18(1), 39; https://doi.org/10.3390/v18010039 - 25 Dec 2025
Viewed by 433
Abstract
Parvovirus B19 (B19V) is a human ssDNA virus with ample pathogenic potential. It is characterized by a selective tropism for erythroid progenitor cells (EPC), exerting a cytotoxic effect with blockade of erythropoiesis. In our work, we investigated both viral and cellular expression profile [...] Read more.
Parvovirus B19 (B19V) is a human ssDNA virus with ample pathogenic potential. It is characterized by a selective tropism for erythroid progenitor cells (EPC), exerting a cytotoxic effect with blockade of erythropoiesis. In our work, we investigated both viral and cellular expression profile in the course of infection of EPCs cultures via mRNA high throughput sequencing technology (HTS) and a dedicated bioinformatic pipeline, reconstructing both the viral and cellular transcriptome and their variations. A productive infection was confirmed as restricted to EPCs expressing mature differentiation markers and the specific receptor for virus VP1u region. mRNA HTS reconstructed the viral transcriptome in terms of localization and abundance of the different mRNA species, detailing the differential expression profile of B19V among early or late times in the course of infection. Analysis of cellular transcriptome indicated that variation was mainly driven by the cellular differentiation process, with the virus impacting to a lesser level, but still clearly separating infected vs. non-infected profiles. At early times post-infection, variations were typical of cellular sensing of viral infection and aimed at the induction of an antiviral state. At later times in the course of infection, the cellular population showed induction of an inflammatory response, related to TNF and IL-10, and a transition to adaptive immunity with evidence of upregulation of genes involved in MHC-II presentation. This dual-transcriptome analysis on infected EPCs population can lay the ground for future research aimed at a better definition of the pathogenetic mechanisms of B19V. Full article
(This article belongs to the Collection Parvoviridae)
Show Figures

Figure 1

24 pages, 1274 KB  
Review
The Non-Coding RNome Landscape in Erythropoiesis: Pathophysiological Implications
by Emma Brisot, Laurent Metzinger and Valérie Metzinger-Le Meuth
Cells 2025, 14(24), 1971; https://doi.org/10.3390/cells14241971 - 11 Dec 2025
Viewed by 373
Abstract
Erythropoiesis is a multistage process critical for red blood cell production and systemic oxygen transport. It is tightly regulated, and recent advances have highlighted the pivotal regulatory roles of non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), in governing both [...] Read more.
Erythropoiesis is a multistage process critical for red blood cell production and systemic oxygen transport. It is tightly regulated, and recent advances have highlighted the pivotal regulatory roles of non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), in governing both physiological and pathological erythropoiesis. These ncRNAs have roles in the fine-tuning of the classical transcriptional and post-transcriptional control. This review explores the complex landscape of the non-coding RNome in erythroid differentiation, maturation, and function. We summarize how specific miRNAs influence erythroid lineage commitment, hemoglobin switching, iron metabolism, and cellular morphology, as well as their modulation by environmental and pathological cues. We also discuss emerging evidence on lncRNAs regulating chromatin remodeling, alternative splicing, apoptosis, enucleation, and erythroid-specific gene expression. These insights suggest that ncRNAs are instrumental orchestrators of erythropoiesis and accordingly, potential biomarkers and therapeutic targets in anemia and related hematologic disorders. Full article
Show Figures

Figure 1

22 pages, 1103 KB  
Review
Traditional Health Practices May Promote Nrf2 Activation Similar to Exercise
by Hubert Kolb, Stephan Martin and Kerstin Kempf
Int. J. Mol. Sci. 2025, 26(23), 11546; https://doi.org/10.3390/ijms262311546 - 28 Nov 2025
Viewed by 1369
Abstract
Various non-pharmacological practices have been reported to enhance overall health. The molecular effects of exercise have been shown to involve the upregulation of enzymes and transcription factors that enhance antioxidative and anti-inflammatory activity, boost mitochondrial function and growth, and promote a parasympathetic tone. [...] Read more.
Various non-pharmacological practices have been reported to enhance overall health. The molecular effects of exercise have been shown to involve the upregulation of enzymes and transcription factors that enhance antioxidative and anti-inflammatory activity, boost mitochondrial function and growth, and promote a parasympathetic tone. These beneficial changes occur as an adaptive/hormetic response to an initial increase in oxygen radical and nitric oxide production in working muscles. The redox-sensitive nuclear factor erythroid 2-related factor 2 (Nrf2) was identified as the key mediator of the cellular defense response. A similar adaptive response appears to occur in response to exposure to heat or cold, hyperbaric or hypobaric oxygen, cupping therapy, acupuncture, caloric restriction, and the consumption of polyphenol-rich plant-based foods or spices, and there is direct or indirect evidence for the involvement of Nrf2. In many cases, additional stress signaling pathways have been observed to be upregulated, including the nicotinamide adenine dinucleotide (NAD+)-sirtuin and the adenosine monophosphate (AMP)-activated protein kinase pathways. We conclude that while several traditional health practices may share a hormetic mechanism—mild radical-induced damage triggers a defense response through upregulation of antioxidative, anti-inflammatory, and repair activities, which may impact body-wide tissue function. Full article
(This article belongs to the Special Issue Role of NRF2 Pathway in Chronic Diseases)
Show Figures

Figure 1

23 pages, 2166 KB  
Review
Dimethyl Fumarate vs. Monomethyl Fumarate: Unresolved Pharmacologic Issues
by Jana Kopincova and Iveta Bernatova
Pharmaceutics 2025, 17(12), 1506; https://doi.org/10.3390/pharmaceutics17121506 - 22 Nov 2025
Viewed by 927
Abstract
Dimethyl fumarate (DMF) has established a significant position among therapies for multiple sclerosis and psoriasis and is now being investigated for repurposing to many other non-malignant diseases. Despite decades of preclinical research, some issues about its pharmacology remain unresolved, with ongoing debate over [...] Read more.
Dimethyl fumarate (DMF) has established a significant position among therapies for multiple sclerosis and psoriasis and is now being investigated for repurposing to many other non-malignant diseases. Despite decades of preclinical research, some issues about its pharmacology remain unresolved, with ongoing debate over which of the methyl esters of fumarate, whether DMF or monomethyl fumarate (MMF), is the active ingredient. It is generally accepted that DMF undergoes enzymatic hydrolysis to MMF and methanol. However, there is disagreement regarding its exact site, its extent, and the responsible enzyme(s). The enzymatic mechanisms, particularly the roles of carboxylesterases-1 and 2, vary across tissues and species, complicating the translation of in vitro and in vivo preclinical findings into clinical practice. In addition, the impact of DMF and MMF is often not clearly distinguishable and sometimes overlaps, making the true molecular mediators of therapeutic and side effects unclear. Thus, the interpretation of some results obtained in studies is inconsistent because of interchanging of in vitro and in vivo observed features of fumarate esters: while DMF demonstrates rapid and strong effects in cell culture studies, including nuclear factor erythroid 2-related factor 2 (NRF2) function activation and glutathione depletion, these observations may not exactly reflect systemic pharmacology and physiology dominated by MMF. Moreover, methanol, the co-product of DMF metabolism, may contribute to the observed DMF effects through increased production of reactive oxygen species, which could result in activation of NRF2-dependent mechanisms. This review highlights specific unresolved issues in DMF metabolism, which are sometimes overlooked. Full article
Show Figures

Graphical abstract

16 pages, 3591 KB  
Review
Residual Genetic Material in Mature Red Blood Cells
by Georgios Dryllis, Sotirios P. Fortis, Aspasia Kouroupaki, Ioannis Tsamesidis, Vassilios Birtsas, Andreas G. Tsantes, Serena Valsami, Konstantinos Konstantopoulos, Effie G. Papageorgiou, Ilias Pessach and Anastasios G. Kriebardis
Int. J. Mol. Sci. 2025, 26(21), 10774; https://doi.org/10.3390/ijms262110774 - 5 Nov 2025
Viewed by 1865
Abstract
Mature erythrocytes are traditionally regarded as anucleate cells lacking nuclear DNA. However, evidence shows they retain residual genetic material, including mitochondrial DNA (mtDNA) and RNA fragments. This review explores the role of such genetic material in cellular function, diagnostics, and erythropoiesis. A comprehensive [...] Read more.
Mature erythrocytes are traditionally regarded as anucleate cells lacking nuclear DNA. However, evidence shows they retain residual genetic material, including mitochondrial DNA (mtDNA) and RNA fragments. This review explores the role of such genetic material in cellular function, diagnostics, and erythropoiesis. A comprehensive literature review was conducted, focusing on (i) erythropoiesis, (ii) enucleation of erythroid precursors, (iii) the presence of DNA in red blood cells (RBCs), and (iv) RNA fragments such as messenger RNA (mRNA), microRNA (miRNA), and other non-coding RNAs. Mature RBCs harbor small amounts of DNA and diverse RNA species. Residual DNA can act as damage-associated molecular patterns (DAMPs), triggering immune responses when released under stress or injury. RNA fragments reflect the transcriptional activity of precursor cells and have been linked to potential diagnostic applications. Studies suggest that RBC-derived RNA signatures may serve as non-invasive biomarkers for diseases such as diabetes, cardiovascular conditions, and hematological disorders. These profiles mirror changes in erythropoiesis and provide insights into systemic pathophysiology. Residual genetic material in RBCs extends their role beyond oxygen transport. It contributes to immune modulation and may provide novel diagnostic and therapeutic opportunities, enhancing disease detection and understanding of erythropoiesis. Full article
Show Figures

Figure 1

21 pages, 2821 KB  
Article
High-Frequency, Low-Intensity Pulsed Electric Field and N-Acetylcysteine Synergistically Protect SH-SY5Y Cells Against Hydrogen Peroxide-Induced Cell Damage In Vitro
by Fang-Tzu Hsu, Yu-Yi Kuo and Chih-Yu Chao
Antioxidants 2025, 14(10), 1267; https://doi.org/10.3390/antiox14101267 - 21 Oct 2025
Viewed by 1201
Abstract
Oxidative stress plays an important role in the progression of neurodegenerative diseases (NDDs), and N-acetylcysteine (NAC) has gained attention as a potential agent due to its antioxidant capabilities. This study investigated the synergistic neuroprotective effects of combining NAC with non-contact, high-frequency, low-intensity pulsed [...] Read more.
Oxidative stress plays an important role in the progression of neurodegenerative diseases (NDDs), and N-acetylcysteine (NAC) has gained attention as a potential agent due to its antioxidant capabilities. This study investigated the synergistic neuroprotective effects of combining NAC with non-contact, high-frequency, low-intensity pulsed electric field (H-LIPEF) stimulation on SH-SY5Y human neuronal cells subjected to hydrogen peroxide (H2O2)-induced oxidative damage. It was found that after SH-SY5Y cells were pretreated with NAC and exposed to H-LIPEF stimulation, the oxidative stress of cells was reduced in the subsequent treatment with H2O2. The results showed that the combined NAC and H-LIPEF treatment significantly improved cell viability and more effectively reduced mitochondrial apoptosis. Mechanistic analyses revealed that the combination substantially decreased levels of superoxide and intracellular H2O2, which was associated with enhanced activation of the phosphorylated Akt (p-Akt)/nuclear factor erythroid 2-related factor 2 (Nrf2)/superoxide dismutase type 2 (SOD2) signaling pathway. Furthermore, the treatment reduced the accumulation of 8-oxo-2′-deoxyguanosine triphosphate (8-oxo-dG) accumulation and elevated MutT homolog 1 (MTH1) expression, indicating a protective effect against oxidative DNA damage. These results suggest that H-LIPEF enhances the neuroprotective efficacy of low-dose NAC, highlighting the potential of this combination approach as a new therapeutic strategy for the treatment of NDDs. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

17 pages, 1775 KB  
Article
AI-Driven Analysis for Real-Time Detection of Unstained Microscopic Cell Culture Images
by Kathrin Hildebrand, Tatiana Mögele, Dennis Raith, Maria Kling, Anna Rubeck, Stefan Schiele, Eelco Meerdink, Avani Sapre, Jonas Bermeitinger, Martin Trepel and Rainer Claus
AI 2025, 6(10), 271; https://doi.org/10.3390/ai6100271 - 18 Oct 2025
Viewed by 1460
Abstract
Staining-based assays are widely used for cell analysis but are invasive, alter physiology, and prevent longitudinal monitoring. Label-free, morphology-based approaches could enable real-time, non-invasive drug testing, yet detection of subtle and dynamic changes has remained difficult. We developed a deep learning framework for [...] Read more.
Staining-based assays are widely used for cell analysis but are invasive, alter physiology, and prevent longitudinal monitoring. Label-free, morphology-based approaches could enable real-time, non-invasive drug testing, yet detection of subtle and dynamic changes has remained difficult. We developed a deep learning framework for stain-free monitoring of leukemia cell cultures using automated bright-field microscopy in a semi-automated culture system (AICE3, LABMaiTE, Augsburg, Germany). YOLOv8 models were trained on images from K562, HL-60, and Kasumi-1 cells, using an NVIDIA DGX A100 GPU for training and tested on GPU and CPU environments for real-time performance. Comparative benchmarking with RT-DETR and interpretability analyses using Eigen-CAM and radiomics (RedTell) was performed. YOLOv8 achieved high accuracy (mAP@0.5 > 98%, precision/sensitivity > 97%), with reproducibility confirmed on an independent dataset from a second laboratory and an AICE3 setup. The model distinguished between morphologically similar leukemia lines and reliably classified untreated versus differentiated K562 cells (hemin-induced erythroid and PMA-induced megakaryocytic; >95% accuracy). Incorporation of decitabine-treated cells demonstrated applicability to drug testing, revealing treatment-specific and intermediate phenotypes. Longitudinal monitoring captured culture- and time-dependent drift, enabling separation of temporal from drug-induced changes. Radiomics highlighted interpretable features such as size, elongation, and texture, but with lower accuracy than the deep learning approach. To our knowledge, this is the first demonstration that deep learning resolves subtle, drug-induced, and time-dependent morphological changes in unstained leukemia cells in real time. This approach provides a robust, accessible framework for label-free longitudinal drug testing and establishes a foundation for future autonomous, feedback-driven platforms in precision oncology. Ultimately, this approach may also contribute to more precise and adaptive clinical decision-making, advancing the field of personalized medicine. Full article
(This article belongs to the Special Issue AI in Bio and Healthcare Informatics)
Show Figures

Figure 1

16 pages, 4613 KB  
Article
Inhibitory Effect of S0859 on the Antioxidant Master Switch Nuclear Factor Erythroid 2-Related Factor 2 in Lung Cancer Cells
by Eunsun Lee and Jeong Hee Hong
Antioxidants 2025, 14(10), 1191; https://doi.org/10.3390/antiox14101191 - 28 Sep 2025
Viewed by 919
Abstract
Cancer cells possess endogenous antioxidant systems such as nuclear factor erythroid 2-related factor 2 (NRF2). The electroneutral sodium bicarbonate cotransporter NBCn1, known as a migratory module, is closely associated with cancer metastasis; however, its regulatory signaling in cancer remains unclear. In particular, the [...] Read more.
Cancer cells possess endogenous antioxidant systems such as nuclear factor erythroid 2-related factor 2 (NRF2). The electroneutral sodium bicarbonate cotransporter NBCn1, known as a migratory module, is closely associated with cancer metastasis; however, its regulatory signaling in cancer remains unclear. In particular, the regulation of NBCn1 in response to oxidative stress and its relationship with NRF2 need to be elucidated. In the present study, we found that hydrogen peroxide–induced oxidative stress dysregulated NBCn1 via inhibition of NF-κB, thereby suppressing cellular migration in non-small cell lung cancer A549 cells. Phosphorylation of NF-κB was required for maintaining NBCn1 function in A549 cells. Oxidative stress also induced NRF2 nuclear translocation, reduced NBC activity, and activated oxidative stress–responsive gene expression. Treatment with the NBC inhibitor S0859 impaired ERK activation, NRF2 nuclear translocation, and oxidative stress defense gene expression in A549 cells. Furthermore, oxidative stimulation in the presence of S0859 disrupted the NRF2-mediated oxidative stress defense system and cellular migration in A549 lung cancer cells. Collectively, these findings suggest that S0859, as a potential NRF2 inhibitor, may exert anti-cancer properties. Full article
(This article belongs to the Special Issue Oxidative Stress and NRF2 in Health and Disease—2nd Edition)
Show Figures

Figure 1

14 pages, 1960 KB  
Article
TP53 Expression Status Alters Hemoglobinization and Ferroptosis Sensitivity in K-562 Cells
by Cameron Cardona, Madelyne Young and McKale Montgomery
Int. J. Mol. Sci. 2025, 26(17), 8359; https://doi.org/10.3390/ijms26178359 - 28 Aug 2025
Viewed by 1203
Abstract
Activation of TP53 signaling during ribosome biogenesis is an essential part of erythroid development, whereas the pathologic activation of TP53 in ribosomopathies such as Diamond-Blackfan anemia (DBA) and del (5q) myelodysplastic syndrome (MDS) prevents the normal expansion of erythroid precursors. TP53 can also [...] Read more.
Activation of TP53 signaling during ribosome biogenesis is an essential part of erythroid development, whereas the pathologic activation of TP53 in ribosomopathies such as Diamond-Blackfan anemia (DBA) and del (5q) myelodysplastic syndrome (MDS) prevents the normal expansion of erythroid precursors. TP53 can also be linked to the pathogenesis of DBA and MDS via ferroptosis, a form of iron-mediated cell death propagated by excess polyunsaturated fatty acid-containing oxidizable phospholipids and loss of lipid peroxide repair capacity. The primary objective of this work was to establish how overexpression and mutation of the TP53 gene influences lipid composition, erythroid differentiation, and ferroptosis sensitivity in K-562 cells, an in vitro model for studying erythropoiesis. Employing a reverse genetics approach, we generated four isogenic cell lines that either lacked functional TP53 expression, expressed wild-type (WT) TP53, or expressed one of the two most common TP53 mutation types, R175H or R282W. We then utilized non-targeted lipidomics to quantify and identify changes in specific lipid species that occur with induction of WT and mutant TP53 expression. We also analyzed differences in gene expression, ferroptosis sensitivity, and hemoglobinization by qPCR, CCK-8 cytotoxicity assay, and o-dianisidine staining, respectively. The abundance of 337 distinct lipid species was impacted by induction of WT TP53 expression compared to K-562 cells expressing a nonfunctional P53 protein. Yet only 17 lipid compounds were differentially impacted between cells expressing WT TP53 and either of the mutant TP53 genes tested. Similarly, while the TP53 null K-562 cells displayed modest sensitivity to ferroptosis, cells expressing both WT and mutant TP53 genes were remarkably resistant to ferroptosis. However, terminal differentiation and hemoglobinization were significantly impacted in R175H mutant TP53-expressing K-562 cells. Findings from this work provide novel insights into the role of TP53 in lipid metabolism and terminal erythropoiesis. Full article
(This article belongs to the Special Issue Lipidomics and Lipid Metabolism in Health and Disease)
Show Figures

Figure 1

35 pages, 2019 KB  
Review
Non-Electrophilic Activation of NRF2 in Neurological Disorders: Therapeutic Promise of Non-Pharmacological Strategies
by Chunyan Li, Keren Powell, Luca Giliberto, Christopher LeDoux, Cristina d’Abramo, Daniel Sciubba and Yousef Al Abed
Antioxidants 2025, 14(9), 1047; https://doi.org/10.3390/antiox14091047 - 25 Aug 2025
Cited by 2 | Viewed by 3282
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) serves as a master transcriptional regulator of cellular antioxidant responses through orchestration of cytoprotective gene expression, establishing its significance as a therapeutic target in cerebral pathophysiology. Classical electrophilic NRF2 activators, despite potent activation potential, exhibit paradoxically [...] Read more.
Nuclear factor erythroid 2-related factor 2 (NRF2) serves as a master transcriptional regulator of cellular antioxidant responses through orchestration of cytoprotective gene expression, establishing its significance as a therapeutic target in cerebral pathophysiology. Classical electrophilic NRF2 activators, despite potent activation potential, exhibit paradoxically reduced therapeutic efficacy relative to single antioxidants, attributable to concurrent oxidative stress generation, glutathione depletion, mitochondrial impairment, and systemic toxicity. Although emerging non-electrophilic pharmacological activators offer therapeutic potential, their utility remains limited by bioavailability and suboptimal potency, underscoring the imperative for innovative therapeutic strategies to harness this cytoprotective pathway. Non-pharmacological interventions, including neuromodulation, physical exercise, and lifestyle modifications, activate NRF2 through non-canonical, non-electrophilic pathways involving protein–protein interaction inhibition, KEAP1 degradation, post-translational and transcriptional modulation, and protein stabilization, though mechanistic characterization remains incomplete. Such interventions utilize multi-mechanistic approaches that synergistically integrate multiple non-electrophilic NRF2 pathways or judiciously combine electrophilic and non-electrophilic mechanisms while mitigating electrophile-induced toxicity. This strategy confers neuroprotective effects without the contraindications characteristic of classical electrophilic activators. This review comprehensively examines the mechanistic underpinnings of non-pharmacological NRF2 modulation, highlighting non-electrophilic activation pathways that bypass the limitations inherent to electrophilic activators. The evidence presented herein positions non-pharmacological interventions as viable therapeutic approaches for achieving non-electrophilic NRF2 activation in the treatment of cerebrovascular and neurodegenerative pathologies. Full article
(This article belongs to the Special Issue Oxidative Stress and NRF2 in Health and Disease—2nd Edition)
Show Figures

Figure 1

13 pages, 1294 KB  
Review
VEXAS Syndrome: Genetics, Gender Differences, Clinical Insights, Diagnostic Pitfalls, and Emerging Therapies
by Salvatore Corrao, Marta Moschetti, Salvatore Scibetta, Luigi Calvo, Annarita Giardina, Ignazio Cangemi, Carmela Zizzo, Paolo Colomba and Giovanni Duro
Int. J. Mol. Sci. 2025, 26(16), 7931; https://doi.org/10.3390/ijms26167931 - 17 Aug 2025
Viewed by 2777
Abstract
VEXAS syndrome (Vacuoles, E1-enzyme, X-linked, Autoinflammation, and Somatic) is a recently identified late-onset autoinflammatory disorder characterized by a unique interplay between hematological and inflammatory manifestations. It results from somatic mutations in the UBA1 gene, located on the short arm of the X chromosome. [...] Read more.
VEXAS syndrome (Vacuoles, E1-enzyme, X-linked, Autoinflammation, and Somatic) is a recently identified late-onset autoinflammatory disorder characterized by a unique interplay between hematological and inflammatory manifestations. It results from somatic mutations in the UBA1 gene, located on the short arm of the X chromosome. Initially, females were considered mere carriers, with the syndrome primarily affecting males over 50. However, recent evidence indicates that heterozygous females can exhibit symptoms as severe as those seen in hemizygous males. The disease manifests as systemic inflammation, macrocytic anemia, thrombocytopenia, chondritis, neutrophilic dermatoses, and steroid-dependent inflammatory symptoms. Due to its overlap with autoimmune and hematologic disorders such as relapsing polychondritis, Still’s disease, and myelodysplastic syndromes, misdiagnosis is common. At the molecular level, VEXAS syndrome is driven by impaired ubiquitination pathways, resulting in dysregulated immune responses and clonal hematopoiesis. A key diagnostic marker is the presence of cytoplasmic vacuoles in myeloid and erythroid precursors, though definitive diagnosis requires genetic testing for UBA1 mutations. Traditional immunosuppressants and TNF inhibitors are generally ineffective, while JAK inhibitors and IL-6 blockade provide partial symptom control. Azacitidine and decitabine have shown promise in reducing disease burden, but hematopoietic stem cell transplantation (HSCT) remains the only curative treatment, albeit with significant risks. This review provides a comprehensive analysis of VEXAS syndrome, examining its clinical features, differential diagnoses, diagnostic challenges, and treatment approaches, including both pharmacological and non-pharmacological strategies. By enhancing clinical awareness and optimizing therapeutic interventions, this article aims to bridge emerging genetic insights with practical patient management, ultimately improving outcomes for those affected by this complex and often life-threatening disease. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

64 pages, 7848 KB  
Review
A Reassessment of Sarcopenia from a Redox Perspective as a Basis for Preventive and Therapeutic Interventions
by Alessia Arcaro, Alessio Lepore, Giovanni Paolo Cetrangolo, Gianluca Paventi, Paul Richard Julian Ames and Fabrizio Gentile
Int. J. Mol. Sci. 2025, 26(16), 7787; https://doi.org/10.3390/ijms26167787 - 12 Aug 2025
Cited by 3 | Viewed by 2862
Abstract
The use of a wide variety of antioxidants has been advocated as a means to prevent, delay the progression of, or counteract the adverse consequences of sarcopenia, such as loss of muscle strength, muscle quantity/quality, and physical performance. However, these proposals do not [...] Read more.
The use of a wide variety of antioxidants has been advocated as a means to prevent, delay the progression of, or counteract the adverse consequences of sarcopenia, such as loss of muscle strength, muscle quantity/quality, and physical performance. However, these proposals do not always appear to be supported in the literature by a thorough understanding of the contribution of redox perturbations to the pathogenesis of sarcopenia, nor of the biochemical properties, mechanism of action, pharmacokinetics, and pharmacodynamics of different antioxidants. This review discusses these aspects, aiming to provide a rationale for the selection and use of antioxidants in sarcopenia. After providing a definition of sarcopenia in the context of frailty, we distinguish between oxidative eustress as a physiological response of muscle cells to mild stimulation, such as moderate exercise, mediating their capacity for adaptation and regeneration, and oxidative distress as a pathophysiological response to muscle cell damage and death. The role of oxidative damage to biological macromolecules, both direct and mediated by advanced lipid peroxidation end products and advanced glycation/glycoxidation end products, is examined in detail. Next, we discuss antioxidant defense mechanisms, both enzymatic and non-enzymatic, including redox-sensitive gene regulatory events presided over by nuclear factor erythroid 2-related factor 2, the master regulator of enzymatic antioxidants. The review then discusses criteria for a rational classification of non-enzymatic antioxidants. This is followed by a review of some of the main radical-trapping antioxidants, both phenolic and non-phenolic, whose characteristics are compared. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Back to TopTop