Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (161)

Search Parameters:
Keywords = chromatin and nuclear organization

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 2584 KB  
Hypothesis
New Roles of bZIP-Containing Membrane-Bound Transcription Factors in Chromatin Tethering and Karyoptosis
by Dohyun Jeung, Xianzhe Li and Yong-Yeon Cho
Int. J. Mol. Sci. 2025, 26(22), 10896; https://doi.org/10.3390/ijms262210896 - 10 Nov 2025
Viewed by 140
Abstract
The nuclear membrane has emerged as a dynamic regulatory platform coordinating genome organization, mechanotransduction, and regulated cell death (RCD). Beyond its barrier function, the nuclear skeleton—comprising lamins, actin–myosin isoforms, nuclear matrix proteins, and the LINC complex—supports nuclear integrity and gene regulation. Recent evidence [...] Read more.
The nuclear membrane has emerged as a dynamic regulatory platform coordinating genome organization, mechanotransduction, and regulated cell death (RCD). Beyond its barrier function, the nuclear skeleton—comprising lamins, actin–myosin isoforms, nuclear matrix proteins, and the LINC complex—supports nuclear integrity and gene regulation. Recent evidence shows that type II membrane-bound bZIP transcription factors such as cAMP-responsive element-binding protein 3 (CREB3) and CREB3L1 localize to the inner nuclear membrane (INM), linking chromatin tethering with stress signaling. Their stress-induced cleavage by S1P/S2P disrupts chromatin anchoring and, in some contexts, triggers karyoptosis, a novel form of RCD defined by nuclear rupture. These findings position the nuclear envelope (NE) as a mechanosensitive signaling hub with direct implications for disease and therapy. In this review, we provide a comprehensive discussion on how type II membrane-bound bZIP transcription factors and chromatin acting as a nucleoskeleton cooperate to regulate nuclear membrane integrity. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

13 pages, 1832 KB  
Article
Reversine-Induced Telomere Architecture Remodeling in Chronic Myeloid Leukemia Cell Lines: Insights from TeloView® Analysis of 3D Nuclear Architecture
by Fábio Morato de Oliveira, Isabela Dias Cruvinel, Bruno Machado Rezende Ferreira and Sabine Mai
Curr. Issues Mol. Biol. 2025, 47(11), 907; https://doi.org/10.3390/cimb47110907 - 31 Oct 2025
Viewed by 283
Abstract
Reversine is a small-molecule Aurora kinase inhibitor known for its pro-apoptotic effects and potential to remodel chromatin architecture. Although its impact on mitotic regulation is established, its effects on telomere dynamics and nuclear organization in chronic myeloid leukemia (CML) remain unclear. This study [...] Read more.
Reversine is a small-molecule Aurora kinase inhibitor known for its pro-apoptotic effects and potential to remodel chromatin architecture. Although its impact on mitotic regulation is established, its effects on telomere dynamics and nuclear organization in chronic myeloid leukemia (CML) remain unclear. This study aimed to investigate the effects of reversine on telomere architecture, genomic instability, and apoptosis in CML cell lines (K-562 and MEG-01). Reversine was applied at increasing concentrations, and cytotoxicity was assessed using caspase-3/7 activation assays. Quantitative PCR was used to measure AURKA and AURKB mRNA expressions. Three-dimensional telomere architecture was analyzed with TeloView® v1.03 software after Q-FISH labeling to quantify telomere number, signal intensity, aggregation, nuclear volume, and a/c ratio. Reversine induced a dose- and time-dependent apoptotic response in both cell lines and significantly downregulated AURKA and AURKB expressions. Three-dimensional telomere analysis revealed a marked reduction in telomere number and aggregates, signal intensity, and nuclear volume. While reduced signal intensity may indicate telomere shortening, the concurrent decrease in aggregation and altered spatial parameters suggests telomeric reorganization rather than progressive instability. These features reflect structural nuclear remodeling and early apoptotic commitment. Differences between K-562 and MEG-01 responses underscore potential heterogeneity in telomere maintenance mechanisms. Reversine modulates genomic stability in CML cells through dual mechanisms involving Aurora kinase inhibition and telomere architecture remodeling. The integration of 3D telomere profiling highlights reversine’s potential as a therapeutic agent targeting nuclear disorganization and mitotic dysregulation in leukemia. Full article
(This article belongs to the Special Issue Cancer Biomarkers: Discovery and Applications)
Show Figures

Figure 1

15 pages, 700 KB  
Review
The Ca2+ Bridge: From Neurons to Circuits in Rett Syndrome
by Luis Molina Calistro, Yennyfer Arancibia, Javiera Alarcón and Rodrigo Flavio Torres
Int. J. Mol. Sci. 2025, 26(21), 10490; https://doi.org/10.3390/ijms262110490 - 29 Oct 2025
Viewed by 374
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder caused primarily by mutations in the gene encoding the methyl-CpG-binding protein 2 (Mecp2). Mecp2 binds to methylated cytosines, playing a crucial role in chromatin organization and transcriptional regulation. At the neurobiological level, RTT is characterized [...] Read more.
Rett syndrome (RTT) is a severe neurodevelopmental disorder caused primarily by mutations in the gene encoding the methyl-CpG-binding protein 2 (Mecp2). Mecp2 binds to methylated cytosines, playing a crucial role in chromatin organization and transcriptional regulation. At the neurobiological level, RTT is characterized by dendritic spine dysgenesis and altered excitation–inhibition balance, drawing attention to the mechanisms that scale from mutations in a nuclear protein to altered neuronal connectivity. Although Mecp2 dysfunction disrupts multiple neuronal processes, emerging evidence highlights altered calcium (Ca2+) signaling as a central contributor to RTT pathophysiology. This review explores the link between Mecp2 and Ca2+ regulation by highlighting how Mecp2 affects Ca2+-dependent transcriptional pathways, while Ca2+ modulates Mecp2 function by inducing post-translational modifications. We discuss this crosstalk in light of evidence from RTT models, with a particular focus on the brain-derived neurotrophic factor BDNF-miR132-Mecp2 axis and the dysregulation of ryanodine receptors (RyRs). Additionally, we examine how these perturbations contribute to the reduced structural plasticity and the altered activity-driven gene expression that characterizes RTT. Understanding the intersection between Mecp2 function and Ca2+ homeostasis will provide critical insights into RTT pathogenesis and potential therapeutic targets aimed at restoring neuronal connectivity. Full article
(This article belongs to the Special Issue Calcium Homeostasis of Cells in Health and Disease: Third Edition)
Show Figures

Figure 1

22 pages, 1118 KB  
Review
The Biological Function of Genome Organization
by Xin Yang, Hongni Zhu, Yajie Liu, Jinhong Wang, Yi Song, Shasha Liao and Peng Dong
Int. J. Mol. Sci. 2025, 26(18), 9058; https://doi.org/10.3390/ijms26189058 - 17 Sep 2025
Viewed by 1174
Abstract
The mammalian genome is hierarchically packaged into distinct functional units, including chromatin loops, topologically associating domains, compartments and chromosome territories. This structural organization is fundamentally important because it orchestrates essential nuclear functions that underpin normal cellular identity and organismal development. In this review, [...] Read more.
The mammalian genome is hierarchically packaged into distinct functional units, including chromatin loops, topologically associating domains, compartments and chromosome territories. This structural organization is fundamentally important because it orchestrates essential nuclear functions that underpin normal cellular identity and organismal development. In this review, we synthesize current understanding of the intricate relationship between genome architecture and its critical biological roles. We discuss how hierarchical structures are dynamically established and maintained by architectural proteins, transcription factors, epigenetic regulators and non-coding RNAs via distinct mechanisms. Importantly, we focus on the functional consequences of three-dimensional (3D) genome organization and discuss how it modulates fundamental biological processes such as transcription, gene co-expression, epigenetic modification, DNA replication and repair. We also examine the dynamics of 3D genome organization during cellular differentiation, early embryonic development and organogenesis, followed by discussing how structural disruptions are mechanistically linked to various human diseases. Understanding the biological function of 3D genome organization is thus not only essential for deciphering fundamental nuclear processes but also holds significant promise for elucidating disease etiologies and developing effective therapeutics. Full article
(This article belongs to the Special Issue Recent Advances in Chromatin Structure and Dynamics)
Show Figures

Figure 1

45 pages, 3161 KB  
Review
Drosophila as a Model for Studying the Roles of Lamins in Normal Tissues and Laminopathies
by Aleksandra Zielińska, Marta Rowińska, Aleksandra Tomczak and Ryszard Rzepecki
Cells 2025, 14(17), 1303; https://doi.org/10.3390/cells14171303 - 22 Aug 2025
Viewed by 1287
Abstract
Nuclear processes are fundamental to the regulation of cellular, tissue, and organismal function, especially in complex multicellular systems. Central to these processes are lamins and lamin-associated proteins, which contribute to nuclear structure, gene expression, and chromatin organization. The discovery that mutations in genes [...] Read more.
Nuclear processes are fundamental to the regulation of cellular, tissue, and organismal function, especially in complex multicellular systems. Central to these processes are lamins and lamin-associated proteins, which contribute to nuclear structure, gene expression, and chromatin organization. The discovery that mutations in genes coding for lamins and lamina-associated proteins give rise to rare disorders—collectively called laminopathies—has intensified interest in this field among cell biologists and medical scientists. While many practical and clinically relevant questions about phenotype development and potential treatments require mammalian models, key molecular mechanisms and interactions have also been effectively studied in both vertebrate and invertebrate systems. This review focuses on a discussion of Drosophila lamins, their major properties, functions, interactions and post-translational modifications, with comparison to mammalian lamins, and a discussion of the value of fly models in studies of lamins in muscle tissue development and function in comparison to mammalian lamin B-type and A/C-type. In this paper, we have discussed the overall impact of lamin Dm and lamin C level manipulations on overall phenotype, especially on larval and adult muscles. We have thoroughly discussed the conclusions, which may have been drawn from experiments with overexpression of lamin C mutants mimicking lamin A laminopathy mutations. We have presented and discussed the suggestion that the mechanisms underlying Drosophila muscle phenotype development are similar not only to human dystrophic laminopathies but also to classical human muscular dystrophies such as Duchenne muscular dystrophy and Hutchison–Gilford Progeria syndrome. We suggest that the activation of the stress response contributes to the laminopathic phenotype detected in Drosophila. Finely, this review discusses in depth the lamin Dm and lamin C interactomes, discrepancies between String-based interactome networks, and our map of interactomes based on manual verification of experimental data on Drosophila lamin interactions. Full article
(This article belongs to the Section Cellular Biophysics)
Show Figures

Figure 1

20 pages, 1716 KB  
Review
Ferroptosis as a Form of Cell Death—Medical Importance and Pharmacological Implications
by Blanka Kielan, Artur Pałasz, Krzysztof Krysta and Marek Krzystanek
Pharmaceuticals 2025, 18(8), 1183; https://doi.org/10.3390/ph18081183 - 11 Aug 2025
Viewed by 1242
Abstract
Background/Objectives: Ferroptosis is a regulated form of cell death that occurs in the state of oxidative–antioxidative imbalance of an organism. The main components of ferroptosis are lipid peroxidation and iron accumulation. Cells experiencing ferroptosis show swelling, shrunken mitochondria with an abnormal structure, atrophic [...] Read more.
Background/Objectives: Ferroptosis is a regulated form of cell death that occurs in the state of oxidative–antioxidative imbalance of an organism. The main components of ferroptosis are lipid peroxidation and iron accumulation. Cells experiencing ferroptosis show swelling, shrunken mitochondria with an abnormal structure, atrophic cristae, dense mitochondrial membranes, and ruptured outer membrane. Ferroptotic cells demonstrate a normal nucleus size without nuclear concentration, and neither condensation nor chromatin margination. Ferroptosis is regulated by multiple protein, genetic, and metabolic factors. The aim of this article is to present ferroptosis as a model of cell death occurring in various conditions and diseases. Methods: A literature search of PubMed, Web of Science was performed. Search terms included “ferroptosis”, “lipid peroxidation”, “iron”, and “cell death”. Results: Ferroptosis affects the onset, course, progression, and treatment of diseases, including neurodegenerative diseases, cancer diseases, autoimmune diseases, and hemorrhages. By using appropriate ferroptosis moderators, it is possible to influence the course of the disease in patients. Conclusions: By understanding the ferroptosis phenomenon well, it is possible to regulate its occurrence by considering the action of oxidative and antioxidant factors. A comprehensive understanding of ferroptosis and the factors regulating this process should be the goal in therapy for many diseases. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Graphical abstract

28 pages, 2248 KB  
Review
Role of Ionizing Radiation in Shaping the Complex Multi-Layered Epigenome
by Claudia E. Rübe, Mutaz A. Abd Al-razaq, Carola Meier, Markus Hecht and Christian Rübe
Epigenomes 2025, 9(3), 29; https://doi.org/10.3390/epigenomes9030029 - 8 Aug 2025
Viewed by 1325
Abstract
The impact of ionizing radiation (IR) with induction of various DNA damage is based not only on genetic but also on epigenetic effects. Epigenetic modifications determine the chromatin structure and DNA accessibility, thereby regulating cellular functions through the expression of individual genes or [...] Read more.
The impact of ionizing radiation (IR) with induction of various DNA damage is based not only on genetic but also on epigenetic effects. Epigenetic modifications determine the chromatin structure and DNA accessibility, thereby regulating cellular functions through the expression of individual genes or entire groups of genes. However, the influence of DNA repair processes on the restoration of local chromatin structures and global nuclear architectures is still insufficiently understood. In multicellular organisms, epigenetic mechanisms control diverse cellular functions of specific cell types through precise temporal and spatial regulation of gene expression and silencing. How altered epigenetic mechanisms regulate the pathophysiological function of cells, tissues, and ultimately entire organs following IR exposure remains to be investigated in detail. Radiation-induced epigenetic processes are particularly critical for immature cell populations such as tissue-specific stem and progenitor cells during development and differentiation of organ tissues. Genome-wide patterns of DNA and histone modifications are established cell types—specifically during the development and differentiation of organ tissues but can also be fundamentally altered in adult organism by stress responses, such as radiation-induced DNA damage. Following IR exposure, epigenetic factors are not always fully restored to their original state, resulting in epigenetic dysfunction that causes cells to lose their original identity and function. Moreover, severe radiation-induced DNA damage can induce premature senescence of cells in complex tissues, which ultimately leads to signs of aging and age-related diseases such as cancer. In this work, we provide an overview of the most important epigenetic changes following IR exposure and their pathophysiological significance for the development of acute and chronic radiation reactions. Full article
(This article belongs to the Special Issue Features Papers in Epigenomes 2025)
Show Figures

Figure 1

23 pages, 5695 KB  
Article
Impact of miR-181a on SIRT1 Expression and Senescence in Hutchinson–Gilford Progeria Syndrome
by Eva-Maria Lederer, Felix Quirin Fenzl, Peter Krüger, Moritz Schroll, Ramona Hartinger and Karima Djabali
Diseases 2025, 13(8), 245; https://doi.org/10.3390/diseases13080245 - 4 Aug 2025
Viewed by 702
Abstract
Background/Objectives: Hutchinson–Gilford progeria syndrome (HGPS) is a rare and fatal genetic disease caused by a silent mutation in the LMNA gene, leading to the production of progerin, a defective prelamin A variant. Progerin accumulation disrupts nuclear integrity, alters chromatin organization, and drives systemic [...] Read more.
Background/Objectives: Hutchinson–Gilford progeria syndrome (HGPS) is a rare and fatal genetic disease caused by a silent mutation in the LMNA gene, leading to the production of progerin, a defective prelamin A variant. Progerin accumulation disrupts nuclear integrity, alters chromatin organization, and drives systemic cellular dysfunction. While autophagy and inflammation are key dysregulated pathways in HGPS, the role of microRNAs (miRNAs) in these processes remains poorly understood. Methods: We performed an extensive literature review to identify miRNAs involved in autophagy and inflammation. Through stem-loop RT-qPCR in aging HGPS and control fibroblast strains, we identified significant miRNAs and focused on the most prominent one, miR-181a-5p, for in-depth analysis. We validated our in vitro findings with miRNA expression studies in skin biopsies from an HGPS mouse model and conducted functional assays in human fibroblasts, including immunofluorescence staining, β-Galactosidase assay, qPCR, and Western blot analysis. Transfection studies were performed using an miR-181a-5p mimic and its inhibitor. Results: We identified miR-181a-5p as a critical regulator of premature senescence in HGPS. miR-181a-5p was significantly upregulated in HGPS fibroblasts and an HGPS mouse model, correlating with Sirtuin 1 (SIRT1) suppression and induction of senescence. Additionally, we demonstrated that TGFβ1 induced miR-181a-5p expression, linking inflammation to miRNA-mediated senescence. Inhibiting miR-181a-5p restored SIRT1 levels, increased proliferation, and alleviated senescence in HGPS fibroblasts, supporting its functional relevance in disease progression. Conclusions: These findings highlight the important role of miR-181a-5p in premature aging and suggest its potential as a therapeutic target for modulating senescence in progeroid syndromes. Full article
(This article belongs to the Section Rare Syndrome)
Show Figures

Figure 1

23 pages, 1080 KB  
Review
Epigenetic and Genotoxic Mechanisms of PFAS-Induced Neurotoxicity: A Molecular and Transgenerational Perspective
by Narimane Kebieche, Seungae Yim, Claude Lambert and Rachid Soulimani
Toxics 2025, 13(8), 629; https://doi.org/10.3390/toxics13080629 - 26 Jul 2025
Viewed by 2263
Abstract
Per- and polyfluoroalkyl substances (PFAS) are persistent environmental pollutants that continue to raise concern owing to their ability to accumulate in living organisms. In recent years, a growing body of research has shown that PFAS can exert their toxicity through disruption of both [...] Read more.
Per- and polyfluoroalkyl substances (PFAS) are persistent environmental pollutants that continue to raise concern owing to their ability to accumulate in living organisms. In recent years, a growing body of research has shown that PFAS can exert their toxicity through disruption of both DNA integrity and epigenetic regulation. This includes changes in DNA methylation patterns, histone modifications, chromatin remodeling, and interference with DNA repair mechanisms. These molecular-level alterations can impair transcriptional regulation and cellular homeostasis, contributing to genomic instability and long-term biological dysfunction. In neural systems, PFAS exposure appears particularly concerning. It affects key regulators of neurodevelopment, such as BDNF, synaptic plasticity genes, and inflammatory mediators. Importantly, epigenetic dysregulation extends to non-coding RNAs (ncRNAs), including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), which mediate post-transcriptional silencing and chromatin remodeling. Although direct evidence of transgenerational neurotoxicity is still emerging, animal studies provide compelling hints. Persistent changes in germline epigenetic profiles and transcriptomic alterations suggest that developmental reprogramming might be heritable by future generations. Additionally, PFAS modulate nuclear receptor signaling (e.g., PPARγ), further linking environmental cues to chromatin-level gene regulation. Altogether, these findings underscore a mechanistic framework in which PFAS disrupt neural development and cognitive function via conserved epigenetic and genotoxic mechanisms. Understanding how these upstream alterations affect long-term neurodevelopmental and neurobehavioral outcomes is critical for improving risk assessment and guiding future interventions. This review underscores the need for integrative research on PFAS-induced chromatin disruptions, particularly across developmental stages, and their potential to impact future generations. Full article
(This article belongs to the Special Issue PFAS Toxicology and Metabolism—2nd Edition)
Show Figures

Figure 1

11 pages, 1654 KB  
Article
Binding of Transcriptional Activator to Silent Loci Causes Their Detachment from the Nuclear Lamina in Drosophila Neurons and Salivary Gland Cells
by Ruslan A. Simonov, Oxana M. Olenkina, Valentina V. Nenasheva, Yuri A. Abramov, Sergey A. Lavrov, Anna A. Fedotova and Yuri Y. Shevelyov
Int. J. Mol. Sci. 2025, 26(12), 5793; https://doi.org/10.3390/ijms26125793 - 17 Jun 2025
Viewed by 951
Abstract
In mammals, the binding of transcriptional activators leads to the repositioning of silent loci from the nuclear periphery to the nuclear interior. However, it remained unknown whether the same mechanism functions in Drosophila. Here, using FISH and DamID, we have shown that [...] Read more.
In mammals, the binding of transcriptional activators leads to the repositioning of silent loci from the nuclear periphery to the nuclear interior. However, it remained unknown whether the same mechanism functions in Drosophila. Here, using FISH and DamID, we have shown that binding the GAL4 activator to the silent loci causes weakening of their interactions with the nuclear lamina and relocalization inside nuclei in Drosophila salivary gland cells and neurons. This mimics the removal from the nuclear periphery of a neuron-specific gene upon its activation in neurons. Salivary gland cells contain polytene chromosomes with mechanical properties, different from chromosomes of diploid cells, while neurons represent predominantly non-dividing cell type. Our results indicate a causal relationship between transcriptional activator binding and changes in the intranuclear position of loci in Drosophila. They also point to the similarity in general chromatin dynamics in mammals and Drosophila, thus strengthening the role of model organisms in studying genome architecture. Full article
(This article belongs to the Special Issue Drosophila: A Versatile Model in Biology and Medicine—2nd Edition)
Show Figures

Figure 1

21 pages, 5057 KB  
Article
Genetic Factors Linking Nucleolar Stress with R2 Retrotransposon Expression in Drosophila melanogaster
by Shova Pandey, An Tri Nguyen, Audrey K. Maricle and Patrick J. DiMario
Int. J. Mol. Sci. 2025, 26(12), 5480; https://doi.org/10.3390/ijms26125480 - 7 Jun 2025
Viewed by 946
Abstract
R2 retrotransposons reside exclusively within the 28S regions of 10–20% of all rDNA genes comprising the nucleolar organizer loci on the X and Y chromosomes of Drosophila melanogaster. These R2-inserted genes are normally silent and heterochromatic. When expressed, however, the R2 [...] Read more.
R2 retrotransposons reside exclusively within the 28S regions of 10–20% of all rDNA genes comprising the nucleolar organizer loci on the X and Y chromosomes of Drosophila melanogaster. These R2-inserted genes are normally silent and heterochromatic. When expressed, however, the R2 transcript is co-transcribed with the 28S rRNA. Self-cleavage releases a 3.6 kb mature R2 transcript that encodes a single protein with endonuclease and reverse transcriptase activities that facilitate R2 element transposition by target-primed reverse transcription. While we know the molecular details of R2 transposition, we know little about the genetic mechanisms that initiate R2 transcription. Here, we examine R2 expression in wild type versus mutant backgrounds. R2 expression in stage 1–4 wild type egg chambers was variable depending on the stock. R2 expression was silent in wild type stages 5–10 but was consistently active during nurse cell nuclear breakdown in stages 12–13 regardless of the genetic background. Massive R2 expression occurred in stages 5–10 upon loss of Udd, an RNA Pol I transcription factor. Similarly, loss of Nopp140, an early ribosome assembly factor, induced R2 expression more so in somatic tissues. Interestingly, over-expression of the Nopp140-RGG isoform but not the Nopp140-True isoform induced R2 expression in larval somatic tissues, suggesting Nopp140-RGG could potentially affect rDNA chromatin structure. Conversely, Minute mutations in genes encoding ribosomal proteins had minor positive effects on R2 expression. We conclude that R2 expression is largely controlled by factors regulating RNA Pol I transcription and early ribosome assembly. Full article
(This article belongs to the Special Issue Modulation of Transcription: Imag(in)ing a Fundamental Mechanism)
Show Figures

Figure 1

41 pages, 28708 KB  
Article
Identification and Characterization of LINE and SINE Retrotransposons in the African Hedgehog (Atelerix albiventris, Erinaceidae) and Their Association with 3D Genome Organization and Gene Expression
by Mengyuan Zhu, Jianxuan Zhou, Nannan Chen, Jianing Xu, Haipeng Wang, Libo Jiang and Fengtang Yang
Genes 2025, 16(4), 397; https://doi.org/10.3390/genes16040397 - 29 Mar 2025
Viewed by 1592
Abstract
Background: The African hedgehog (Atelerix albiventris) exhibits specialized skin differentiation leading to spine formation, yet its regulatory mechanisms remain unclear. Transposable elements (TEs), particularly LINEs (long interspersed nuclear elements) and SINEs (short interspersed nuclear elements), are known to influence genome organization [...] Read more.
Background: The African hedgehog (Atelerix albiventris) exhibits specialized skin differentiation leading to spine formation, yet its regulatory mechanisms remain unclear. Transposable elements (TEs), particularly LINEs (long interspersed nuclear elements) and SINEs (short interspersed nuclear elements), are known to influence genome organization and gene regulation. Objectives: Given the high proportion of SINEs in the hedgehog genome, this study aims to characterize the distribution, evolutionary dynamics, and potential regulatory roles of LINEs and SINEs, focusing on their associations with chromatin architecture, DNA methylation, and gene expression. Methods: We analyzed LINE and SINE distribution using HiFi sequencing and classified TE families through phylogenetic reconstruction. Hi-C data were used to explore TE interactions with chromatin architecture, while whole-genome 5mCpG methylation was inferred from PacBio HiFi reads of muscle tissue using a deep-learning-based approach. RNA-seq data from skin tissues were analyzed to assess TE expression and potential associations with genes linked to spine development. Results: SINEs form distinct genomic blocks in GC-rich and highly methylated regions, whereas LINEs are enriched in AT-rich, hypomethylated regions. LINEs and SINEs are associated differently with A/B compartments, with SINEs in euchromatin and LINEs in heterochromatin. Methylation analysis suggests that younger TEs tend to have higher methylation levels, and expression analysis indicates that some differentially expressed TEs may be linked to genes involved in epidermal and skeletal development. Conclusions: This study provides a genome-wide perspective on LINE and SINE distribution, methylation patterns, and potential regulatory roles in A. albiventris. While not establishing a direct causal link, the findings suggest that TEs may influence gene expression associated with spine development, offering a basis for future functional studies. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

18 pages, 3649 KB  
Review
Interplay and Dynamics of Chromatin Architecture and DNA Damage Response: An Overview
by Susanna Ambrosio, Anna Noviello, Giovanni Di Fusco, Francesca Gorini, Anna Piscone, Stefano Amente and Barbara Majello
Cancers 2025, 17(6), 949; https://doi.org/10.3390/cancers17060949 - 11 Mar 2025
Cited by 1 | Viewed by 2834
Abstract
Genome stability is safeguarded by a finely orchestrated cascade of events that collectively represent the DNA damage response (DDR). In eukaryotes, the DDR operates within the dynamic chromatin landscape, where the interplay between DNA repair factors, chromatin remodeling, replication, transcription, spatial genome organization, [...] Read more.
Genome stability is safeguarded by a finely orchestrated cascade of events that collectively represent the DNA damage response (DDR). In eukaryotes, the DDR operates within the dynamic chromatin landscape, where the interplay between DNA repair factors, chromatin remodeling, replication, transcription, spatial genome organization, and cytoskeletal forces is tightly coordinated. High-resolution studies have unveiled chromatin alterations spanning multiple scales, from localized kilobase-level changes to megabase-scale reorganization, which impact chromatin’s physical properties and enhance the mobility of damaged regions. Leveraging this knowledge could pave the way for innovative therapeutic strategies, particularly in targeting chromatin dynamics to destabilize cancer cells selectively. This review, focusing on DNA double-strand breaks (DSBs), sheds light on how chromatin undergoes dynamic modifications in response to damage and how these changes influence the DDR at both local and global levels, offering a glimpse into how nuclear architecture contributes to the delicate balance between genome stability and adaptability and highlighting the importance of exploring these interactions in the context of cancer therapy. Full article
(This article belongs to the Special Issue Genome Instability and Human Cancer)
Show Figures

Figure 1

17 pages, 3977 KB  
Article
Promyelocytic Leukemia Protein (PML) Regulates Stem Cell Pluripotency Through Novel Sumoylation Targets
by Syrago Spanou, Takis Makatounakis, Chrysa Filippopoulou, Georgios Dougalis, George Stamatakis, Christoforos Nikolaou, Martina Samiotaki, Georgia Chachami, Joseph Papamatheakis and Androniki Kretsovali
Int. J. Mol. Sci. 2025, 26(3), 1145; https://doi.org/10.3390/ijms26031145 - 28 Jan 2025
Viewed by 1773
Abstract
The promyelocytic leukemia protein (PML) and its associated nuclear bodies have recently emerged as critical regulators of embryonic stem (ES) cell identity. Despite their recognized importance, the complete spectrum of PML-mediated molecular events in ES cells remains unclear. In this report, we study [...] Read more.
The promyelocytic leukemia protein (PML) and its associated nuclear bodies have recently emerged as critical regulators of embryonic stem (ES) cell identity. Despite their recognized importance, the complete spectrum of PML-mediated molecular events in ES cells remains unclear. In this report, we study how PML is shaping the proteomic and SUMO proteomic landscape in ES cells. Proteomic profiling of PML-depleted ES cells uncovered a downregulation of self-renewal factors and an upregulation of proteins associated with translation and proteasomal activity, reflecting a cellular transition from pluripotency to differentiation. Importantly, PML promotes the sumoylation of pluripotency-related factors, chromatin organizers, and cell cycle regulators. We identified SALL1 and CDCA8 as novel PML-directed sumoylation targets, both critical for ES cell maintenance. SALL1 sumoylation increases the activation of the Wnt pathway, contributing to its ability to inhibit ES cell differentiation. Similarly, CDCA8 sumoylation enhances its capacity to promote cell proliferation. Collectively, our findings demonstrate that PML regulates ES cell identity by modulating the abundance or sumoylation of key regulators involved in pluripotency and cell cycle progression. Full article
Show Figures

Figure 1

20 pages, 14502 KB  
Article
Homeodomain Involvement in Nuclear HOX Protein Homo- and Heterodimerization
by Damien Marchese, Laetitia Evrard, Isabelle Bergiers, Ludovic Boas, Justine Duphénieux, Maryse Hermant, Tamara Pringels, Fisnik Zeqiri, Marc Pirson, Jean-Claude Twizere, Françoise Gofflot, René Rezsohazy and Laure Bridoux
Int. J. Mol. Sci. 2025, 26(1), 423; https://doi.org/10.3390/ijms26010423 - 6 Jan 2025
Viewed by 3857
Abstract
HOX genes play essential roles in patterning the anteroposterior axis of animal embryos and in the formation of various organs. In mammals, there are 39 HOX genes organized into four clusters (HOXA–D) located on different chromosomes. In relationship with their orderly arrangement along [...] Read more.
HOX genes play essential roles in patterning the anteroposterior axis of animal embryos and in the formation of various organs. In mammals, there are 39 HOX genes organized into four clusters (HOXA–D) located on different chromosomes. In relationship with their orderly arrangement along the chromosomes, these genes show nested expression patterns which imply that embryonic territories co-express multiple HOX genes along the main body axis. Interactomic database entries, as well as a handful of publications, support that some HOX proteins can form homodimers or interact with other HOX proteins. However, the consequences of HOX protein interactions have been poorly investigated and remain largely elusive. In this study, we compiled a repository of all HOX–HOX interactions from available databases, and taking HOXA1, HOXA2, and HOXA5 as examples, we investigated the capacity of HOX proteins to form homo- and heterodimers. We revealed that while the DNA-binding domain, the homeodomain, is not necessary for HOXA1 homodimerization, the nuclear localization of the dimerization is dependent on the homeodomain, particularly the integrity of the third helix of HOXA1. Furthermore, we demonstrated that HOXA1 can influence the localization of HOXA1 when it is deprived of the homeodomain, increasing its abundance in the chromatin-containing fraction. Moreover, HOXA1 nuclear homodimerization occurs independently of the integrity of the hexapeptide and, consequently, of its well-known interactor, the homeodomain protein PBX. These results hint at a potential involvement of dimerization in the complex landscape of HOX regulatory mechanisms. Full article
(This article belongs to the Special Issue Biomolecular Basis of Life Processes)
Show Figures

Figure 1

Back to TopTop