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

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 (3,035)

Search Parameters:
Keywords = cell nucleus

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 2264 KB  
Article
Effect of Estrogen on Sirt1 Signaling in Human Macrophages
by Maria Luisa Barcena, Anne Breiter, Julia Temp, Yury Ladilov and Vera Regitz-Zagrosek
Int. J. Mol. Sci. 2025, 26(17), 8670; https://doi.org/10.3390/ijms26178670 - 5 Sep 2025
Abstract
Cardiovascular diseases are the leading cause of human death worldwide. The role of the female hormone estrogen (E2) in this context is subject of debate. E2 might counteract inflammation by acting on macrophages; however, the underlying cellular mechanisms remain poorly understood. In the [...] Read more.
Cardiovascular diseases are the leading cause of human death worldwide. The role of the female hormone estrogen (E2) in this context is subject of debate. E2 might counteract inflammation by acting on macrophages; however, the underlying cellular mechanisms remain poorly understood. In the current study, we used primary human macrophages to investigate the effects of E2 on the NAD+-dependent deacetylase Sirt1, protein acetylation, and pro-inflammatory phenotype. Male and female primary monocytes from healthy adult individuals were polarized into pro-inflammatory M1 macrophages via treatment with LPS and IFN-γ followed by treatment with E2 for 24 h. While E2 treatment had no effect on the Sirt1 protein expression, it significantly increased the acetylation state of nuclear proteins p53 and Ku70. In addition, E2 increased NFκB-p65 expression exclusively in male M1 macrophages, while TNF-α was reduced in female M1 macrophages following E2 treatment. In male monocyte-like cells, E2 significantly reduced nuclear Sirt1 expression and increased Ku70 acetylation. The current study demonstrated that E2 treatment of human M1 macrophages leads to downregulation of nuclear Sirt1 and hyperacetylation of corresponding nuclear proteins. These molecular changes are associated with an enhancement of the pro-inflammatory phenotype in male primary macrophages, while an attenuation of inflammation was observed in female cells. Full article
(This article belongs to the Special Issue Steroids in Human Disease and Health)
Show Figures

Figure 1

15 pages, 3377 KB  
Article
Swiprosin-1 Negatively Regulates Osteoclast Differentiation and Bone Resorption via Akt/MAPK/NF-κB Pathway and αvβ3 Integrin-Dependent Signaling
by Yoon-Hee Cheon, Sung Chul Kwak, Chong Hyuk Chung, Chang Hoon Lee, Myeung Su Lee and Ju-Young Kim
Int. J. Mol. Sci. 2025, 26(17), 8613; https://doi.org/10.3390/ijms26178613 - 4 Sep 2025
Viewed by 87
Abstract
Swiprosin-1 (SWS1/EFhd2) is a calcium-binding adaptor protein involved in cytoskeletal regulation, but its physiological role in bone homeostasis remains largely undefined. To elucidate its function in osteoclast biology, we examined SWS1 expression and activity during osteoclastogenesis using primary murine bone marrow-derived macrophages, siRNA-mediated [...] Read more.
Swiprosin-1 (SWS1/EFhd2) is a calcium-binding adaptor protein involved in cytoskeletal regulation, but its physiological role in bone homeostasis remains largely undefined. To elucidate its function in osteoclast biology, we examined SWS1 expression and activity during osteoclastogenesis using primary murine bone marrow-derived macrophages, siRNA-mediated knockdown, and SWS1 knockout (KO) mice. SWS1 was predominantly localized to the nucleus in precursor cells and redistributed to the F-actin ring in mature osteoclasts. Receptor activator of nuclear factor-kappa B ligand stimulation significantly downregulated SWS1 mRNA expression. Loss of SWS1 enhanced osteoclast formation, F-actin ring integrity, and bone resorption, accompanied by elevated expression of osteoclastogenic markers. In vivo, male SWS1 KO mice exhibited deteriorated trabecular bone microarchitecture with increased osteoclast numbers. Mechanistically, SWS1 deficiency intensified αvβ3 integrin-associated cytoskeletal signaling and upregulated Akt, MAPK, NF-κB, and PLCγ2 pathways. These results indicate that SWS1 negatively regulates osteoclast differentiation and function by restraining cytoskeletal reorganization and downstream signaling. Collectively, our findings establish SWS1 as a novel modulator of osteoclast activity and a potential therapeutic target for osteolytic bone disorders. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

19 pages, 1270 KB  
Systematic Review
Neuroimmune Mechanisms in Alcohol Use Disorder: Microglial Modulation and Therapeutic Horizons
by Jiang-Hong Ye, Wanhong Zuo, Faraz Chaudhry and Lawrence Chinn
Psychoactives 2025, 4(3), 33; https://doi.org/10.3390/psychoactives4030033 - 4 Sep 2025
Viewed by 217
Abstract
Alcohol Use Disorder (AUD) profoundly impacts individuals and society, driven by neurobiological adaptations that sustain chronicity and relapse. Emerging research highlights neuroinflammation, particularly microglial activation, as a central mechanism in AUD pathology. Ethanol engages microglia—the brain’s immune cells—through key signaling pathways such as [...] Read more.
Alcohol Use Disorder (AUD) profoundly impacts individuals and society, driven by neurobiological adaptations that sustain chronicity and relapse. Emerging research highlights neuroinflammation, particularly microglial activation, as a central mechanism in AUD pathology. Ethanol engages microglia—the brain’s immune cells—through key signaling pathways such as Toll-like receptor 4 (TLR4) and the NLRP3 inflammasome, triggering the release of proinflammatory cytokines (IL-1β, TNF-α, IL-6). These mediators alter synaptic plasticity in addiction-related brain regions, including the ventral tegmental area, nucleus accumbens, amygdala, and lateral habenula, thereby exacerbating cravings, withdrawal symptoms, and relapse risk. Rodent models reveal that microglial priming disrupts dopamine signaling, heightening impulsivity and anxiety-like behaviors. Human studies corroborate these findings, demonstrating increased microglial activation markers in postmortem AUD brains and neuroimaging analyses. Notably, sex differences influence microglial reactivity, complicating AUD’s neuroimmune landscape and necessitating sex-specific research approaches. Microglia-targeted therapies—including minocycline, ibudilast, GLP-1 receptor agonists, and P2X7 receptor antagonists—promise to mitigate neuroinflammation and reduce alcohol intake, yet clinical validation remains limited. Addressing gaps such as biomarker identification, longitudinal human studies, and developmental mechanisms is critical. Leveraging multi-omics tools and advanced neuroimaging can refine microglia-based therapeutic strategies, offering innovative avenues to break the self-sustaining cycle of AUD. Full article
(This article belongs to the Special Issue Feature Papers in Psychoactives)
Show Figures

Figure 1

18 pages, 3539 KB  
Article
Neuro-Genomic Mapping of Cardiac Neurons with Systemic Analysis Reveals Cognitive and Neurodevelopmental Impacts in Congenital Heart Disease
by Abhimanyu Thakur and Raj Kishore
Life 2025, 15(9), 1400; https://doi.org/10.3390/life15091400 - 4 Sep 2025
Viewed by 132
Abstract
Congenital heart disease (CHD) is associated with neurodevelopmental and cognitive impairments, but the underlying molecular mechanisms remain unclear. This study investigated cardiac neuronal genomics in CHD using single-nucleus RNA-sequencing data (GSE203274) from 157,273 cardiac nuclei of healthy donors and patients with hypoplastic left [...] Read more.
Congenital heart disease (CHD) is associated with neurodevelopmental and cognitive impairments, but the underlying molecular mechanisms remain unclear. This study investigated cardiac neuronal genomics in CHD using single-nucleus RNA-sequencing data (GSE203274) from 157,273 cardiac nuclei of healthy donors and patients with hypoplastic left heart syndrome (HLHS), Tetralogy of Fallot (TOF), dilated (DCM), and hypertrophic (HCM) cardiomyopathies. The Uniform Manifold Approximation and Projection (UMAP) clustering identified major cardiac cell types, revealing neuron-specific transcriptional programmes. Neuronal populations showed enriched expression of neurodevelopmental disorder-linked genes (NRXN3, CADM2, ZNF536) and synaptic signalling pathways. CHD cardiac neurons exhibited upregulated markers of cognitive dysfunction (APP, SNCA, BDNF) and neurodevelopment regulators (DNMT1, HCFC1) across subtypes. Cardiomyocyte troponin elevation correlated with neuronal exosome receptor expression (TLR2, LRP1), suggesting intercellular communication. Gene ontology analysis highlighted overlaps between cardiovascular disease pathways and neurodevelopmental disorder signatures in CHD neurons. These findings provide the first neuro-genomic map of cardiac neurons in CHD, linking cardiac pathology to neural outcomes through transcriptional dysregulation. Further, the systemic analysis of clinical findings in CHD further supports the risk of neurodevelopmental impacts. In summary, this study identifies transcriptional dysregulation within cardiac neurons in CHD and, together with a systemic analysis of clinical data, provides molecular evidence linking cardiac pathology to neurodevelopmental and cognitive impairments. Full article
(This article belongs to the Section Genetics and Genomics)
Show Figures

Figure 1

13 pages, 1779 KB  
Article
Nup153 and TPR/Megator Interact with TREX-2 Subunits and Are Essential for TREX-2-Dependent Nuclear Export of hsp70 mRNA in Drosophila
by Yulia Vdovina, Julia Nikolenko, Anastasia Orlova, Anna Glukhova, Maria Kurshakova, Savva Fet, Anna Tvorogova, Pyotr Tyurin-Kuzmin, Anton Golovnin, Sofia Georgieva and Daria Kopytova
Int. J. Mol. Sci. 2025, 26(17), 8595; https://doi.org/10.3390/ijms26178595 - 4 Sep 2025
Viewed by 147
Abstract
The TREX-2 complex is conserved from yeast to humans and is responsible for mRNA export from the nucleus to the cytoplasm. In yeast and humans, the TPR and Nup153 nucleoporins of the nuclear pore complex are involved in TREX-2-dependent mRNA export, but data [...] Read more.
The TREX-2 complex is conserved from yeast to humans and is responsible for mRNA export from the nucleus to the cytoplasm. In yeast and humans, the TPR and Nup153 nucleoporins of the nuclear pore complex are involved in TREX-2-dependent mRNA export, but data on their involvement in this process is rather controversial. In the present work, we have studied the role of TPR and Nup153 in the TREX-2-dependent export of hsp70 mRNA in Drosophila. We have shown that Nup153 and TPR are required for the TREX-2-dependent export of hsp70 mRNA, and their knockdown leads to mRNA accumulation in the cell nucleus. We have also demonstrated that Nup153 knockdown leads to TPR relocation to the nucleoplasm. Both nucleoporins are required for TREX-2 subunits’ association with the nuclear pore. Nup153 depletion leads to the TREX-2 subunits’ relocation from the nuclear pore to the nucleoplasm. The depletion of TPR leads to PCID2 relocation to the nucleoplasm and Xmas-2 retention at the nuclear pore and does not affect ENY2 redistribution. The TREX-2 subunits form several contacts with Nup153 and TPR. Hence, both nucleoporins are involved in the interaction with TREX-2 and TREX-2-dependent export in Drosophila. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

23 pages, 4560 KB  
Article
A Single-Nucleus Transcriptomic Atlas of the Mouse Lumbar Spinal Cord: Functional Implications of Non-Coding RNAs
by Pablo Ruiz-Amezcua, Miguel Nieto Hernández, Javier García Flores, Clara Plaza Alonso, David Reigada, Teresa Muñoz-Galdeano, Eva Vargas, Rodrigo M. Maza, Francisco J. Esteban and Manuel Nieto-Díaz
BioTech 2025, 14(3), 70; https://doi.org/10.3390/biotech14030070 - 3 Sep 2025
Viewed by 414
Abstract
The adult lumbar spinal cord plays a critical role in locomotor control and somatosensory integration, whose transcriptional architecture under physiological conditions has been characterized in various studies with restricted numbers of individuals (up to four). Here, we present an integrative single-nucleus RNA sequencing [...] Read more.
The adult lumbar spinal cord plays a critical role in locomotor control and somatosensory integration, whose transcriptional architecture under physiological conditions has been characterized in various studies with restricted numbers of individuals (up to four). Here, we present an integrative single-nucleus RNA sequencing (snRNA-seq) atlas of the healthy adult mouse lumbar spinal cord, assembled from over 86,000 nuclei from 16 samples across five public datasets. Using a harmonized computational pipeline, we identify all major spinal cell lineages and resolve 17 transcriptionally distinct neuronal subtypes. A central novelty of our approach is the systematic inclusion of non-coding RNAs (ncRNAs), including long non-coding RNAs (lncRNAs) and pseudogenes. By comparing transcriptomic analyses based on coding-only, non-coding-only, and combined gene sets, we show that ncRNAs, despite accounting to a 10% of the recorded information of each cell, contribute to cell type-specific signatures. This resource offers a high-resolution, ncRNA-inclusive reference for the adult spinal cord and provides a foundation for future studies on spinal plasticity, injury, and regeneration. Full article
(This article belongs to the Special Issue BioTech: 5th Anniversary)
Show Figures

Figure 1

14 pages, 1197 KB  
Article
Optimization of (Dithioperoxo)thiolate-Based Antifungal Agents for Triazole-Resistant Aspergillus Fumigatus
by Surya Karuturi, Kaitlyn L. Jobe, Melinda E. Varney, Michael D. Hambuchen, A. R. M. Ruhul Amin and Timothy E. Long
Pathogens 2025, 14(9), 878; https://doi.org/10.3390/pathogens14090878 - 3 Sep 2025
Viewed by 266
Abstract
This investigation on novel antifungal agents featuring a thiol-reactive (dithioperoxo)thiolate chemical nucleus [-NC(S)S-SR] established that the optimal levels of fungal growth inhibition were achieved with thiomethyl-bound derivatives (R = Me). The most efficacious analogs had MIC50/MIC90 values of 2/2 µg/mL [...] Read more.
This investigation on novel antifungal agents featuring a thiol-reactive (dithioperoxo)thiolate chemical nucleus [-NC(S)S-SR] established that the optimal levels of fungal growth inhibition were achieved with thiomethyl-bound derivatives (R = Me). The most efficacious analogs had MIC50/MIC90 values of 2/2 µg/mL and an MIC range of 1 to 2 µg/mL for a ten-member panel of voriconazole-resistant A. fumigatus mutants. Pharmacodynamic studies revealed that the lead (dithioperoxo)thiolates impaired conidial germination and germling development more effectively than voriconazole for the triazole-resistant strain AR-1295. Moreover, glutathione and Cu2+ were shown to have antagonistic interactions, which was attributed to the thiol-reactive, pro-oxidant properties of the (dithioperoxo)thiolates and their metabolic conversion to chelating agents. Cytotoxicity studies further showed that the compounds were less toxic to human fetal kidney cells than squamous carcinoma cells. The collective findings of the investigation indicate that (dithioperoxo)thiolates are effective antifungal agents against A. fumigatus to merit additional research on their therapeutic potential. Full article
Show Figures

Figure 1

17 pages, 1173 KB  
Article
AL-Net: Adaptive Learning for Enhanced Cell Nucleus Segmentation in Pathological Images
by Zhuping Chen, Sheng-Lung Peng, Rui Yang, Ming Zhao and Chaolin Zhang
Electronics 2025, 14(17), 3507; https://doi.org/10.3390/electronics14173507 - 2 Sep 2025
Viewed by 246
Abstract
Precise segmentation of cell nuclei in pathological images is the foundation of cancer diagnosis and quantitative analysis, but blurred boundaries, scale variability, and staining differences have long constrained its reliability. To address this, this paper proposes AL-Net—an adaptive learning network that breaks through [...] Read more.
Precise segmentation of cell nuclei in pathological images is the foundation of cancer diagnosis and quantitative analysis, but blurred boundaries, scale variability, and staining differences have long constrained its reliability. To address this, this paper proposes AL-Net—an adaptive learning network that breaks through these bottlenecks through three innovative mechanisms: First, it integrates dilated convolutions with attention-guided skip connections to dynamically integrate multi-scale contextual information, adapting to variations in cell nucleus morphology and size. Second, it employs self-scheduling loss optimization: during the initial training phase, it focuses on region segmentation (Dice loss) and later switches to a boundary refinement stage, introducing gradient manifold constraints to sharpen edge localization. Finally, it designs an adaptive optimizer strategy, leveraging symbolic exploration (Lion) to accelerate convergence, and switches to gradient fine-tuning after reaching a dynamic threshold to stabilize parameters. On the 2018 Data Science Bowl dataset, AL-Net achieved state-of-the-art performance (Dice coefficient 92.96%, IoU 86.86%), reducing boundary error by 15% compared to U-Net/DeepLab; in cross-domain testing (ETIS/ColonDB polyp segmentation), it demonstrated over 80% improvement in generalization performance. AL-Net establishes a new adaptive learning paradigm for computational pathology, significantly enhancing diagnostic reliability. Full article
(This article belongs to the Special Issue Image Segmentation, 2nd Edition)
Show Figures

Figure 1

22 pages, 6033 KB  
Article
Survivin Is a Central Mediator of Cell Proliferation in HPV-Negative Head and Neck Squamous Cell Carcinoma
by Jing Zhu, Jianhong An, Erqiang Hu, Gregory Rosenblatt, Gabriela Berner, Aadita Roy, Nicole Kawachi, Nitisha Shrivastava, Vikas Mehta, Jeffrey E. Segall, Michael B. Prystowsky and Thomas J. Ow
Cancers 2025, 17(17), 2864; https://doi.org/10.3390/cancers17172864 - 31 Aug 2025
Viewed by 368
Abstract
Background/Objectives: HNSCC is a highly aggressive malignancy marked by the dysregulation of the cell cycle. In HPV HNSCC, mutations in the CDKN2A gene frequently result in the loss of the p16 protein, a key inhibitor of the cyclin D1/CDK4/6 complex. This loss [...] Read more.
Background/Objectives: HNSCC is a highly aggressive malignancy marked by the dysregulation of the cell cycle. In HPV HNSCC, mutations in the CDKN2A gene frequently result in the loss of the p16 protein, a key inhibitor of the cyclin D1/CDK4/6 complex. This loss results in unchecked G1/S phase progression. The CDK4/6 inhibitor palbociclib has shown therapeutic potential in HPV HNSCC by inducing G1 phase arrest and reducing cell viability. In this study, we investigated the molecular mechanisms by which palbociclib affects cell viability in HPV HNSCC. Methods: Four HPV HNSCC cell lines were treated with palbociclib, and RNA sequencing was performed to assess changes in gene expression. Cell viability was measured using the MTT assay. To further investigate protein localization, interactions, and function, we used immunofluorescence staining, co-immunoprecipitation, small molecule inhibitors, and siRNA-mediated knockdown. Results: We demonstrate that palbociclib downregulates survivin, a protein that plays dual roles in mitosis and apoptosis, thereby inhibiting cell proliferation. We also found that survivin is overexpressed in HPV HNSCC. Inhibiting survivin dimerization using the compound LQZ-7i significantly reduces cell viability and promotes its export from the nucleus to the cytoplasm. Additionally, we identified USP1, a deubiquitinase, as both a downstream target of CDK4/6 and a key regulator of survivin stability. Inhibiting USP1 activity or silencing its expression significantly reduces survivin levels. Conclusions: Our findings highlight survivin as a critical mediator of cell proliferation in HPV HNSCC and suggest that targeting the CDK4/6-USP1-survivin axis may offer a promising therapeutic strategy. Full article
(This article belongs to the Special Issue Genetic Alterations and the Tumor Microenvironment)
Show Figures

Figure 1

18 pages, 20873 KB  
Article
Characterizing Microglial Morphology: Methodological Advances in Confocal Imaging and Analysis
by Juan P. Taborda-Bejarano, David B. Nowak, Fernando Chaure, Malika L. Allen, Kathryn A. Blek, Stephen Walterhouse, John R. Mantsch and Constanza Garcia-Keller
Cells 2025, 14(17), 1354; https://doi.org/10.3390/cells14171354 - 30 Aug 2025
Viewed by 466
Abstract
Microglia are central to neuroimmune responses and undergo dynamic structural and functional changes in models of stress and addiction, and in response to pharmacological treatments. While transcriptomic and proteomic assays provide insights into molecular profiles, morphological analysis remains a valuable proxy for assessing [...] Read more.
Microglia are central to neuroimmune responses and undergo dynamic structural and functional changes in models of stress and addiction, and in response to pharmacological treatments. While transcriptomic and proteomic assays provide insights into molecular profiles, morphological analysis remains a valuable proxy for assessing region-specific microglial response. However, morphological features alone often fail to capture the full complexity of microglial function, underscoring the need for standardized methods and complementary approaches. Here, we describe a standardized imaging pipeline for analyzing microglia in the nucleus accumbens core (NAcore), integrating unbiased confocal image acquisition with precise anatomical reference points. We compare two widely used image analysis platforms—IMARIS and CellSelect-3DMorph—highlighting their workflows, output metrics, and utility in quantifying microglial morphology following treatment with adenosine triphosphate (ATP). Both tools detect well described features of microglial dynamics, though they differ in automation level, analysis speed, and output types. Our findings demonstrate that both platforms provide reliable morphological data, with CellSelect-3DMorph offering a rapid, open-access alternative for high-throughput analysis. Additionally, using software-derived parameters in principal component analysis clustering has proven useful for identifying distinct subpopulations of microglia separated by their morphology. This work provides a practical framework for morphological analysis and promotes reproducibility in microglial studies under environmental and pharmacological interventions. Full article
Show Figures

Figure 1

14 pages, 1689 KB  
Article
Crossing Barriers: PEGylated Gold Nanoparticles as Promising Delivery Vehicles for siRNA Delivery in Alzheimer’s Disease
by Elżbieta Okła, Marcin Hołota, Sylwia Michlewska, Serafin Zawadzki, Katarzyna Miłowska, Javier Sánchez-Nieves, Rafael Gómez, Francisco Javier De la Mata, Maria Bryszewska and Maksim Ionov
Biomedicines 2025, 13(9), 2108; https://doi.org/10.3390/biomedicines13092108 - 29 Aug 2025
Viewed by 374
Abstract
Background: The proportion of people suffering from neurodegenerative conditions, such as Alzheimer’s disease (AD), is increasing in the population year on year. Despite the constant effort of researchers, these conditions remain incurable and can only be managed by alleviation or delaying of [...] Read more.
Background: The proportion of people suffering from neurodegenerative conditions, such as Alzheimer’s disease (AD), is increasing in the population year on year. Despite the constant effort of researchers, these conditions remain incurable and can only be managed by alleviation or delaying of symptoms. The lack of suitable treatment is caused by constricted access to the brain, limited by the brain-blood barrier. The aim of this work was to investigate two pegylated gold nanoparticles as potential carriers of therapeutic siRNA and their impact on the cellular functions of Human Brain Endothelial Cells. Methods and Results: Nanoparticles AuNP14a and AuNP14b complexed with siRNA were internalized by HBEC-5i cells and located in the cytoplasm. The genotoxicity assay proved that the nucleus was not affected and complexed nanoparticles did not cause DNA damage. The reactive oxygen species formation and mitochondrial membrane potential changes were measured and showed an adaptive response of cells after compound administration. Results obtained in a cytotoxicity assay conducted on astrocytes and pericytes, which are components of the blood–brain barrier, confirmed the biosafety of tested nanoparticles. Conclusions: In summary, it was shown that AuNP14a and AuNP14b are promising candidates as nanocarriers for therapeutic nucleic acids through biological barriers. Full article
(This article belongs to the Special Issue Recent Advances in Targeted Drug Delivery Systems)
Show Figures

Graphical abstract

14 pages, 2304 KB  
Article
Functional Study of the WRKY Transcription Factor Family PgWRKY064-04 Gene in Panax ginseng
by Mengna Liu, En Yu, Tao Liu, Jiaqing Liu, Lihe Hou, Mingzhu Zhao, Meiping Zhang, Yi Wang, Yue Zhang and Kangyu Wang
Agriculture 2025, 15(17), 1837; https://doi.org/10.3390/agriculture15171837 - 29 Aug 2025
Viewed by 303
Abstract
Ginseng (Panax ginseng) is a valuable medicinal plant whose primary active components, known as ginsenosides, play a significant role in anti-cancer, anti-inflammatory, and anti-diabetic effects. WRKY transcription factors represent a prominent class of transcription factors in higher plants, fulfilling essential functions [...] Read more.
Ginseng (Panax ginseng) is a valuable medicinal plant whose primary active components, known as ginsenosides, play a significant role in anti-cancer, anti-inflammatory, and anti-diabetic effects. WRKY transcription factors represent a prominent class of transcription factors in higher plants, fulfilling essential functions in numerous processes such as plant growth and development, reactions to biotic and abiotic stresses, and the control of secondary metabolism. This study is based on the laboratory’s previous bioinformatics analysis of the WRKY gene family in ginseng. After in-depth analysis, the PgWRKY064-04 gene was identified, which is significantly associated with ginsenosides. The physicochemical properties and expression patterns of this gene were analyzed, indicating that its expression in ginseng is temporally and spatially specific. A subcellular localization vector for this gene was constructed, confirming that it functions in the cell nucleus. Subsequently, overexpression vectors and interference vectors for PgWRKY064-04 were constructed, and ginseng adventitious roots were transformed using Agrobacterium-mediated transformation, successfully yielding positive materials. Gene expression levels and saponin content in the positive materials were detected, preliminary findings indicate that the expression of the PgWRKY064-04 gene is negatively correlated with the biosynthesis of ginsenosides. This study complements research on the functional roles of WRKY transcription factor family genes in ginseng, paving the way for future efforts to enhance ginsenoside production using modern biotechnological approaches. Full article
(This article belongs to the Special Issue Genetic Diversity Assessment and Phenotypic Characterization of Crops)
Show Figures

Figure 1

18 pages, 5537 KB  
Article
Effect of Singlet Oxygen on the Stomatal and Cell Wall of Rice Seedling Under Different Stresses
by Yao Xiao, Zhong-Wei Zhang, Xin-Yue Yang, Lin-Bei Xie, Li-Ping Chen, Yang-Er Chen, Ming Yuan, Guang-Deng Chen and Shu Yuan
Int. J. Mol. Sci. 2025, 26(17), 8382; https://doi.org/10.3390/ijms26178382 - 28 Aug 2025
Viewed by 262
Abstract
Singlet oxygen (1O2), a reactive oxygen species, can oxidize lipids, proteins, and DNA at high concentrations, leading to cell death. Despite its extremely short half-life (10−5 s), 1O2 acts as a critical signaling molecule, triggering a [...] Read more.
Singlet oxygen (1O2), a reactive oxygen species, can oxidize lipids, proteins, and DNA at high concentrations, leading to cell death. Despite its extremely short half-life (10−5 s), 1O2 acts as a critical signaling molecule, triggering a retrograde pathway from chloroplasts to the nucleus to regulate nuclear gene expression. In this study, rice seeds were treated with 0, 5, 20 and 80 μM Rose Bengal (RB, a photosensitizer) under moderate light for 3 days to induce 1O2 generation. Treatment with 20 μM RB reduced stomatal density by approximately 25% in three-leaf-stage rice seedlings, while increasing the contents of pectin, hemicellulose, and cellulose in root cell walls by 30–40%. Under drought, salinity, or shading stress, 20 μM RB treatment significantly improved rice tolerance, as evidenced by higher relative water contents (49–58%) and chlorophyll contents (60–76%) and lower malondialdehyde (37–43%) and electrolyte leakage (29–37%) compared to the control. Moreover, RT-qPCR analysis revealed that the significant up-regulation of stomatal development genes (OsTMM and OsβCA1) and cell wall biosynthesis genes (OsF8H and OsLRX2) was associated with RB-induced 1O2 production. Thus, under controlled environmental conditions, 1O2 may regulate stomatal development and cell wall remodeling to enhance rice tolerance to multiple abiotic stresses. These results provide new perspectives for the improvement of rice stress tolerance. Full article
(This article belongs to the Special Issue Plant Physiology and Molecular Nutrition: 2nd Edition)
Show Figures

Figure 1

21 pages, 2800 KB  
Article
Nuclear Roles of Spliceosome-Associated microRNAs in Neuronal Cancer Cells
by Shelly Mahlab-Aviv, Keren Or Swissa, Maram Arafat, Keren Zohar, Ayelet Rachel Peretz, Michal Linial and Ruth Sperling
Int. J. Mol. Sci. 2025, 26(17), 8349; https://doi.org/10.3390/ijms26178349 - 28 Aug 2025
Viewed by 341
Abstract
MicroRNAs (miRNAs) are well known for regulating translation in the cytoplasm, yet their nuclear roles remain poorly understood. Previously, we identified spliceosome-associated miRNAs implicated in tumorigenesis and metastasis in breast cancer models. Here, we investigate their nuclear functions in the immortalized human cortical [...] Read more.
MicroRNAs (miRNAs) are well known for regulating translation in the cytoplasm, yet their nuclear roles remain poorly understood. Previously, we identified spliceosome-associated miRNAs implicated in tumorigenesis and metastasis in breast cancer models. Here, we investigate their nuclear functions in the immortalized human cortical neuron (HCN) cell line, along with glioblastoma (U87MG) and neuroblastoma (SH-SY5Y) cell lines, both widely used as models for brain cancer research. Our findings reveal that spliceosome-associated miRNAs mark neuronal cancer cells and uncover novel nuclear targets. Notably, some spliceosomal miRNAs exhibit opposing regulatory effects in the nucleus compared to the cytoplasm, while others demonstrate potential novel nuclear functions. A prominent example is miR-99b, which overlaps the 5′ splice junction of the poorly characterized long non-coding RNA (lncRNA) sperm acrosome-associated 6 antisense RNA1 (SPACA6-AS1) and, through base pairing, enhances SPACA6-AS1 pre-mRNA levels. These results highlight the diverse and context-dependent functions of nuclear miRNAs in gene regulation and cancer progression, broadening our understanding of their regulatory potential beyond the cytoplasm. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

14 pages, 268 KB  
Review
Beyond the Eye: Glaucoma and the Brain
by Marco Zeppieri, Federico Visalli, Mutali Musa, Alessandro Avitabile, Rosa Giglio, Daniele Tognetto, Caterina Gagliano, Fabiana D’Esposito and Francesco Cappellani
Brain Sci. 2025, 15(9), 934; https://doi.org/10.3390/brainsci15090934 - 28 Aug 2025
Viewed by 419
Abstract
Glaucoma is traditionally classified as an ocular disease characterized by progressive retinal ganglion cell (RGC) loss and optic nerve damage. However, emerging evidence suggests that its pathophysiology may extend beyond the eye, involving trans-synaptic neurodegeneration along the visual pathway and structural changes within [...] Read more.
Glaucoma is traditionally classified as an ocular disease characterized by progressive retinal ganglion cell (RGC) loss and optic nerve damage. However, emerging evidence suggests that its pathophysiology may extend beyond the eye, involving trans-synaptic neurodegeneration along the visual pathway and structural changes within central brain regions, including the lateral geniculate nucleus and visual cortex. In this narrative review, we have used the phrase ‘brain involvement’ to underscore central changes that accompany or follow retinal ganglion cell loss; we have not intended to redefine glaucoma as a primary cerebral disorder. Neuroimaging studies and neurocognitive assessments in adult glaucoma patients, primarily older individuals with primary open-angle glaucoma reveal that glaucoma patients may exhibit alterations in brain connectivity and cortical thinning, aligning it more closely with neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease. This evolving neurocentric perspective raises important questions regarding shared mechanisms—such as mitochondrial dysfunction, chronic inflammation, and impaired axonal transport—that may link glaucomatous optic neuropathy to central nervous system (CNS) pathology. These insights open promising therapeutic avenues, including the repurposing of neuroprotective and neuroregenerative agents, targeting not only intraocular pressure (IOP) but also broader CNS pathways. Furthermore, neuroimaging biomarkers and brain-targeted interventions may play a future role in diagnosis, prognosis, and individualized treatment. This review synthesizes current evidence supporting glaucoma as a CNS disease, explores the mechanistic overlap with neurodegeneration, and discusses the potential clinical implications of glaucoma within a neuro-ophthalmologic paradigm. Full article
Back to TopTop