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

Article Types

Countries / Regions

Search Results (222)

Search Parameters:
Keywords = histone demethylases

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
38 pages, 2158 KiB  
Review
Epigenetic Modulation and Bone Metastasis: Evolving Therapeutic Strategies
by Mahmoud Zhra, Jasmine Hanafy Holail and Khalid S. Mohammad
Pharmaceuticals 2025, 18(8), 1140; https://doi.org/10.3390/ph18081140 - 31 Jul 2025
Viewed by 470
Abstract
Bone metastasis remains a significant cause of morbidity and diminished quality of life in patients with advanced breast, prostate, and lung cancers. Emerging research highlights the pivotal role of reversible epigenetic alterations, including DNA methylation, histone modifications, chromatin remodeling complex dysregulation, and non-coding [...] Read more.
Bone metastasis remains a significant cause of morbidity and diminished quality of life in patients with advanced breast, prostate, and lung cancers. Emerging research highlights the pivotal role of reversible epigenetic alterations, including DNA methylation, histone modifications, chromatin remodeling complex dysregulation, and non-coding RNA networks, in orchestrating each phase of skeletal colonization. Site-specific promoter hypermethylation of tumor suppressor genes such as HIN-1 and RASSF1A, alongside global DNA hypomethylation that activates metastasis-associated genes, contributes to cancer cell plasticity and facilitates epithelial-to-mesenchymal transition (EMT). Key histone modifiers, including KLF5, EZH2, and the demethylases KDM4/6, regulate osteoclastogenic signaling pathways and the transition between metastatic dormancy and reactivation. Simultaneously, SWI/SNF chromatin remodelers such as BRG1 and BRM reconfigure enhancer–promoter interactions that promote bone tropism. Non-coding RNAs, including miRNAs, lncRNAs, and circRNAs (e.g., miR-34a, NORAD, circIKBKB), circulate via exosomes to modulate the RANKL/OPG axis, thereby conditioning the bone microenvironment and fostering the formation of a pre-metastatic niche. These mechanistic insights have accelerated the development of epigenetic therapies. DNA methyltransferase inhibitors (e.g., decitabine, guadecitabine) have shown promise in attenuating osteoclast differentiation, while histone deacetylase inhibitors display context-dependent effects on tumor progression and bone remodeling. Inhibitors targeting EZH2, BET proteins, and KDM1A are now advancing through early-phase clinical trials, often in combination with bisphosphonates or immune checkpoint inhibitors. Moreover, novel approaches such as CRISPR/dCas9-based epigenome editing and RNA-targeted therapies offer locus-specific reprogramming potential. Together, these advances position epigenetic modulation as a promising axis in precision oncology aimed at interrupting the pathological crosstalk between tumor cells and the bone microenvironment. This review synthesizes current mechanistic understanding, evaluates the therapeutic landscape, and outlines the translational challenges ahead in leveraging epigenetic science to prevent and treat bone metastases. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Graphical abstract

16 pages, 2408 KiB  
Article
Female Mice Lacking LSD1 in Myeloid Cells Are Resistant to Inflammatory Bone Loss
by Kristina Astleford-Hopper, Flavia Saavedra, Peter Bittner-Eddy, Clara Stein, Jennifer Auger, Rachel Clark, Juan E. Abrahante Llorens, Bryce A. Binstadt, Vivek Thumbigere-Math and Kim C. Mansky
Cells 2025, 14(14), 1111; https://doi.org/10.3390/cells14141111 - 19 Jul 2025
Viewed by 360
Abstract
Osteoclasts, which are derived from myeloid precursors, are essential for physiologic bone remodeling but also mediate pathological bone loss in inflammatory diseases such as periodontitis and rheumatoid arthritis. Lysine-specific demethylase (LSD1/KDM1A) is a histone demethylase that modulates the chromatin landscape via demethylation of [...] Read more.
Osteoclasts, which are derived from myeloid precursors, are essential for physiologic bone remodeling but also mediate pathological bone loss in inflammatory diseases such as periodontitis and rheumatoid arthritis. Lysine-specific demethylase (LSD1/KDM1A) is a histone demethylase that modulates the chromatin landscape via demethylation of H3K4me1/2 and H3K9me1/2, thereby regulating the expression of genes essential for deciding cell fate. We previously demonstrated that myeloid-specific deletion of LSD1 (LSD1LysM-Cre) disrupts osteoclast differentiation, leading to enhanced BV/TV under physiological conditions. In this study, we show that LSD1LysM-Cre female mice are similarly resistant to inflammatory bone loss in both ligature-induced periodontitis and K/BxN serum-transfer arthritis models. Bulk RNA-seq of mandibular-derived preosteoclasts from LSD1LysM-Cre mice with ligature-induced periodontitis revealed the upregulation of genes involved in inflammation, lipid metabolism, and immune response. Notably, LSD1 deletion blocked osteoclastogenesis even under TGF-β and TNF co-stimulation, which is an alternative RANKL-independent differentiation pathway. Upregulation of Nlrp3, Hif1α, and Acod1 in LSD1LysM-Cre preosteoclasts suggests that LSD1 is essential for repressing inflammatory and metabolic programs that otherwise hinder osteoclast commitment. These findings establish LSD1 as a critical epigenetic gatekeeper integrating inflammatory and metabolic signals to regulate osteoclast differentiation and bone resorption. Therapeutic inhibition of LSD1 may selectively mitigate inflammatory bone loss while preserving physiological bone remodeling. Full article
Show Figures

Figure 1

28 pages, 2265 KiB  
Review
A Brief Overview of the Epigenetic Regulatory Mechanisms in Plants
by Theodoros Tresas, Ioannis Isaioglou, Andreas Roussis and Kosmas Haralampidis
Int. J. Mol. Sci. 2025, 26(10), 4700; https://doi.org/10.3390/ijms26104700 - 14 May 2025
Cited by 1 | Viewed by 1254
Abstract
Plants continuously adapt to their environments by responding to various intrinsic and extrinsic signals. They face numerous biotic and abiotic stresses such as extreme temperatures, drought, or pathogens, requiring complex regulatory mechanisms to control gene activity and adapt their proteome for survival. Epigenetic [...] Read more.
Plants continuously adapt to their environments by responding to various intrinsic and extrinsic signals. They face numerous biotic and abiotic stresses such as extreme temperatures, drought, or pathogens, requiring complex regulatory mechanisms to control gene activity and adapt their proteome for survival. Epigenetic regulation plays a crucial role in these adaptations, potentially leading to both heritable and non-heritable changes across generations. This process enables plants to adjust their gene expression profiles and acclimate effectively. It is also vital for plant development and productivity, affecting growth, yield, and seed quality, and enabling plants to “remember” environmental stimuli and adapt accordingly. Key epigenetic mechanisms that play significant roles include DNA methylation, histone modification, and ubiquitin ligase complex activity. These processes, which have been extensively studied in the last two decades, have led to a better understanding of the underlying mechanisms and expanded the potential for improving agriculturally and economically important plant traits. DNA methylation is a fundamental process that regulates gene expression by altering chromatin structure. The addition of methyl groups to cytosines by DNA methylases leads to gene suppression, whereas DNA demethylases reverse this effect. Histone modifications, on the other hand, collectively referred to as the “histone code”, influence chromatin structure and gene activity by promoting either gene transcription or gene silencing. These modifications are either recognized, added, or removed by a variety of enzymes that act practically as an environmental memory, having a significant impact on plant development and the responses of plants to environmental stimuli. Finally, ubiquitin ligase complexes, which tag specific histones or regulatory proteins with ubiquitin, are also crucial in plant epigenetic regulation. These complexes are involved in protein degradation and play important roles in regulating various cellular activities. The intricate interplay between DNA methylation, histone modifications, and ubiquitin ligases adds complexity to our understanding of epigenetic regulation. These mechanisms collectively control gene expression, generating a complex and branching network of interdependent regulatory pathways. A deeper understanding of this complex network that helps plants adapt to environmental changes and stressful conditions will provide valuable insights into the regulatory mechanisms involved. This knowledge could pave the way for new biotechnological approaches and plant breeding strategies aimed at enhancing crop resilience, productivity, and sustainable agriculture. Full article
(This article belongs to the Section Molecular Plant Sciences)
Show Figures

Figure 1

37 pages, 4013 KiB  
Review
Demystifying the Role of Histone Demethylases in Colorectal Cancer: Mechanisms and Therapeutic Opportunities
by Yuanbin Liu, Min Huang, Xia Tian and Xiaodong Huang
Curr. Issues Mol. Biol. 2025, 47(4), 267; https://doi.org/10.3390/cimb47040267 - 9 Apr 2025
Viewed by 1012
Abstract
Histone demethylases (HDMs) play a pivotal role in colorectal cancer (CRC) progression through dynamic epigenetic regulation. This review summarizes the role and therapeutic potential of HDM in CRC. HDMs primarily target lysine (K) for demethylation (lysine demethylase, KDM). The KDM family is divided [...] Read more.
Histone demethylases (HDMs) play a pivotal role in colorectal cancer (CRC) progression through dynamic epigenetic regulation. This review summarizes the role and therapeutic potential of HDM in CRC. HDMs primarily target lysine (K) for demethylation (lysine demethylase, KDM). The KDM family is divided into the lysine-specific demethylase family and the Jumonji C domain-containing family. HDMs play complex roles in CRC cell proliferation, invasion, migration, stemness, epithelial–mesenchymal transition, immune response, and chemoresistance through epigenetic regulation of different histone demethylation sites. Increasing evidence suggests that KDM may interact with certain factors and regulate CRC tumorigenesis by modulating multiple signaling pathways and affecting the transcription of target genes. These processes may be regulated by upstream genes and thus form a complex epigenetic regulatory network. However, the potential roles and regulatory mechanisms of some HDMs in CRC remain understudied. Preclinical studies have revealed that small-molecule inhibitors targeting HDM impact the activity of specific genes and pathways by inhibiting specific HDM expression, thereby reshaping the tumorigenic landscape of CRC. However, the clinical translational potential of these inhibitors remains unexplored. In conclusion, HDMs play a complex and critical role in CRC progression by dynamically regulating histone methylation patterns. These HDMs shape the malignant behavior of CRC by influencing the activity of key pathways and target genes through epigenetic reprogramming. Targeting HDM may be a promising direction for CRC treatment. Further exploration of the role of specific HDMs in CRC and the therapeutic potential of HDM-specific inhibitors is needed in the future. Full article
(This article belongs to the Special Issue Advances in Molecular Pathogenesis Regulation in Cancer 2025)
Show Figures

Figure 1

17 pages, 4661 KiB  
Article
Identification and Characterization of Histone Modification Gene Families and Their Expression Patterns During Pod and Seed Development in Peanut
by Yingying Chang, Yohannes Gelaye, Ruonan Yao, Ping Yang, Jihua Li, Nian Liu, Li Huang, Xiaojing Zhou, Weigang Chen, Bolun Yu, Huifang Jiang, Boshou Liao, Yong Lei and Huaiyong Luo
Int. J. Mol. Sci. 2025, 26(6), 2591; https://doi.org/10.3390/ijms26062591 - 13 Mar 2025
Viewed by 642
Abstract
Histone methylation and acetylation play potential roles in plant growth and development through various histone modification (HM) genes. However, studies of HM genes are still limited in peanut (Arachis hypogaea L.), a globally important oilseed crop. Here, comprehensive identification and investigation of [...] Read more.
Histone methylation and acetylation play potential roles in plant growth and development through various histone modification (HM) genes. However, studies of HM genes are still limited in peanut (Arachis hypogaea L.), a globally important oilseed crop. Here, comprehensive identification and investigation of HM genes were performed using the whole genome of peanut, and a total of 207 AhHMs encoding 108 histone methyltransferases, 51 histone demethylases, 16 histone acetylases, and 32 histone deacetylases were identified. Detailed analysis of these AhHMs, including chromosome locations, gene structures, protein motifs, and protein–protein interactions, was performed. Tandem, segmental, transposed, dispersed, and whole-genome duplications were involved in the evolution and expansion of the HM gene families in peanut. Ka/Ks analysis indicated that the AhHMs underwent purifying selection. The expression profiles of the 207 AhHMs were investigated during the pod and seed development stages on the basis of the transcriptome sequencing results. Quantitative real-time PCR confirmed that eight AhHMs were differentially expressed during pod and seed development. These results provide data support for further studying the epigenetic mechanism of peanut histones, deepen the understanding of seed development, and provide a new direction for the cultivation of more high-yield and high-quality peanut varieties. Full article
(This article belongs to the Special Issue Molecular and Epigenetic Regulation in Seed Development)
Show Figures

Figure 1

24 pages, 4335 KiB  
Article
Exploring Epigenetic Modifiers in Cowpea: Genomic and Transcriptomic Insights into Histone Methyltransferases and Histone Demethylases
by Jéssica Barbara Vieira Viana, José Ribamar Costa Ferreira-Neto, Eliseu Binneck, Roberta Lane de Oliveira Silva, Antônio Félix da Costa and Ana Maria Benko-Iseppon
Stresses 2025, 5(1), 13; https://doi.org/10.3390/stresses5010013 - 13 Feb 2025
Viewed by 682
Abstract
Histone methyltransferases (SDGs) and demethylases (JMJs) are well-established regulators of transcriptional responses in plants under adverse conditions. This study characterized SDG and JMJ enzymes in the cowpea (Vigna unguiculata) genome and analyzed their expression patterns under various stress conditions, including root [...] Read more.
Histone methyltransferases (SDGs) and demethylases (JMJs) are well-established regulators of transcriptional responses in plants under adverse conditions. This study characterized SDG and JMJ enzymes in the cowpea (Vigna unguiculata) genome and analyzed their expression patterns under various stress conditions, including root dehydration and mechanical injury followed by CABMV or CPSMV inoculation. A total of 47 VuSDG genes were identified in the cowpea genome and classified into seven distinct classes: I, II, III, IV, V, VI, and VII. Additionally, 26 VuJMJ-coding genes were identified and categorized into their respective groups: Jmj-only, JMJD6, KDM3, KDM5, and KDM4. Analysis of gene expansion mechanisms for the studied loci revealed a predominance of dispersed duplication and WGD/segmental duplication events, with Ka/Ks ratios indicating that all WGD/segmental duplications are under purifying selection. Furthermore, a high degree of conservation was observed for these loci across species, with legumes displaying the highest conservation rates. Cis-Regulatory Element analysis of VuSDG and VuJMJ gene promoters revealed associations with Dof-type and bZIP transcription factors, both of which are known to play roles in plant stress responses and developmental processes. Differential expression patterns were observed for VuSDG and VuJMJ genes under the studied stress conditions, with the highest number of upregulated transcripts detected during the root dehydration assay. Our expression data suggest that as the referred stress persists, the tolerant cowpea accession decreases methylation activity on target histones and shifts towards enhanced demethylation. This dynamic balance between histone methylation and demethylation may regulate the expression of genes linked to dehydration tolerance. During the mechanical injury and viral inoculation assays, VuSDG and VuJMJ transcripts were upregulated exclusively within 60 min after the initial mechanical injury combined with CABMV or CPSMV inoculation, indicating an early role for these enzymes in the plant’s defense response to pathogen exposure. The current study presents a detailed analysis of histone modifiers in cowpea and indicates their role as important epigenetic regulators modulating stress tolerance. Full article
(This article belongs to the Collection Feature Papers in Plant and Photoautotrophic Stresses)
Show Figures

Figure 1

15 pages, 2992 KiB  
Article
Conditional Knockout Kdm2a Reveals Crucial Involvement in Development and Function of Kidney Collecting Ducts
by Xianrong Xiong, Hailing Yu, Xupeng Li, Yuan Li, Ruilan Zeng, Yufan Wang, Chunhai Zhang, Yan Xiong, Wei Fu, Honghong He, Shi Yin and Jian Li
Int. J. Mol. Sci. 2025, 26(3), 1230; https://doi.org/10.3390/ijms26031230 - 30 Jan 2025
Viewed by 968
Abstract
Lysine-specific histone demethylase 2 (Kdm2a) is essential for histone modifications involved in development and associated diseases. Nevertheless, the specific functions of Kdm2a in renal development and pathology remain largely unexplored. This study aimed to elucidate the roles of Kdm2a in sustaining [...] Read more.
Lysine-specific histone demethylase 2 (Kdm2a) is essential for histone modifications involved in development and associated diseases. Nevertheless, the specific functions of Kdm2a in renal development and pathology remain largely unexplored. This study aimed to elucidate the roles of Kdm2a in sustaining the biological functions of the kidney by generating mutant mice with Kdm2a deletion using the Aqp2-cre/Loxp system. Our findings showed that Kdm2a is widely expressed across various mouse tissues, with particularly high expression in the kidney’s cortex and medulla, surpassing that in other tissues. Despite no observable effects on morphology or survival following the conditional knockout of Kdm2a, there was a significant reduction in body weight and bilateral kidney weight compared to controls, most pronounced at the 5-week-old stage (p < 0.05). Post Kdm2a deletion, kidney metabolic functions were impaired, evidenced by altered levels of creatinine, urea, total cholesterol, and low-density lipoprotein. Histological examination revealed that Kdm2a-null kidneys exhibited signs of dysfunction, characterized by macrophage infiltration, fibrosis, inflammatory cell infiltration, and mild thrombosis. Further studies revealed that the expression of chemokine- and pro-inflammatory cytokine-related genes Il-6, Il-8, Tnf-a, and Il-1β was significantly increased in the kidneys of Kdm2a cKO mice compared with controls (p < 0.05). Additionally, the expression of reabsorption-related genes (Aqp-3, Aqp-5, and Aqp-8) was markedly downregulated in Kdm2a-deficient kidneys compared with controls (p < 0.05). Collectively, these findings suggest that Kdm2a is crucial for maintaining kidney function and development, partly through the suppression of inflammation and regulation of gene expression. However, the underlying molecular mechanisms of Kdm2a in kidney development warrant further investigation. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

34 pages, 6616 KiB  
Review
Homocysteine Metabolites, Endothelial Dysfunction, and Cardiovascular Disease
by Hieronim Jakubowski and Łukasz Witucki
Int. J. Mol. Sci. 2025, 26(2), 746; https://doi.org/10.3390/ijms26020746 - 16 Jan 2025
Cited by 10 | Viewed by 3612
Abstract
Atherosclerosis is accompanied by inflammation that underlies cardiovascular disease (CVD) and its vascular manifestations, including acute stroke, myocardial infarction, and peripheral artery disease, the leading causes of morbidity/mortality worldwide. The monolayer of endothelial cells formed on the luminal surface of arteries and veins [...] Read more.
Atherosclerosis is accompanied by inflammation that underlies cardiovascular disease (CVD) and its vascular manifestations, including acute stroke, myocardial infarction, and peripheral artery disease, the leading causes of morbidity/mortality worldwide. The monolayer of endothelial cells formed on the luminal surface of arteries and veins regulates vascular tone and permeability, which supports vascular homeostasis. Endothelial dysfunction, the first step in the development of atherosclerosis, is caused by mechanical and biochemical factors that disrupt vascular homeostasis and induce inflammation. Together with increased plasma levels of low-density lipoprotein (LDL), diabetes, hypertension, cigarette smoking, infectious microorganisms, and genetic factors, epidemiological studies established that dysregulated metabolism of homocysteine (Hcy) causing hyperhomocysteinemia (HHcy) is associated with CVD. Patients with severe HHcy exhibit severe CVD and die prematurely due to vascular complications. Biochemically, HHcy is characterized by elevated levels of Hcy and related metabolites such as Hcy-thiolactone and N-Hcy-protein, seen in genetic and nutritional deficiencies in Hcy metabolism in humans and animals. The only known source of Hcy in humans is methionine released in the gut from dietary protein. Hcy is generated from S-adenosylhomocysteine (AdoHcy) and metabolized to cystathionine by cystathionine β-synthase (CBS) and to Hcy-thiolactone by methionyl-tRNA synthetase. Hcy-thiolactone, a chemically reactive thioester, modifies protein lysine residues, generating N-homocysteinylated (N-Hcy)-protein. N-Hcy-proteins lose their normal native function and become cytotoxic, autoimmunogenic, proinflammatory, prothrombotic, and proatherogenic. Accumulating evidence, discussed in this review, shows that these Hcy metabolites can promote endothelial dysfunction, CVD, and stroke in humans by inducing pro-atherogenic changes in gene expression, upregulating mTOR signaling, and inhibiting autophagy through epigenetic mechanisms involving specific microRNAs, histone demethylase PHF8, and methylated histone H4K20me1. Clinical studies, also discussed in this review, show that cystathionine and Hcy-thiolactone are associated with myocardial infarction and ischemic stroke by influencing blood clotting. These findings contribute to our understanding of the complex mechanisms underlying endothelial dysfunction, atherosclerosis, CVD, and stroke and identify potential targets for therapeutic intervention. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Treatment of Cardiovascular Diseases)
Show Figures

Figure 1

35 pages, 1450 KiB  
Review
Epigenetic Properties of Compounds Contained in Functional Foods Against Cancer
by Giulia Casari, Brenda Romaldi, Andrea Scirè, Cristina Minnelli, Daniela Marzioni, Gianna Ferretti and Tatiana Armeni
Biomolecules 2025, 15(1), 15; https://doi.org/10.3390/biom15010015 - 26 Dec 2024
Cited by 7 | Viewed by 2771
Abstract
Epigenetics encompasses reversible and heritable genomic changes in histones, DNA expression, and non-coding RNAs that occur without modifying the nucleotide DNA sequence. These changes play a critical role in modulating cell function in both healthy and pathological conditions. Dysregulated epigenetic mechanisms are implicated [...] Read more.
Epigenetics encompasses reversible and heritable genomic changes in histones, DNA expression, and non-coding RNAs that occur without modifying the nucleotide DNA sequence. These changes play a critical role in modulating cell function in both healthy and pathological conditions. Dysregulated epigenetic mechanisms are implicated in various diseases, including cardiovascular disorders, neurodegenerative diseases, obesity, and mainly cancer. Therefore, to develop innovative therapeutic strategies, research for compounds able to modulate the complex epigenetic landscape of cancer is rapidly surging. Dietary phytochemicals, mostly flavonoids but also tetraterpenoids, organosulfur compounds, and isothiocyanates, represent biologically active molecules found in vegetables, fruits, medicinal plants, and beverages. These natural organic compounds exhibit epigenetic modulatory properties by influencing the activity of epigenetics key enzymes, such as DNA methyltransferases, histone acetyltransferases and deacetylases, and histone methyltransferases and demethylases. Due to the reversibility of the modifications that they induce, their minimal adverse effects, and their potent epigenetic regulatory activity, dietary phytochemicals hold significant promise as antitumor agents and warrant further investigation. This review aims to consolidate current data on the diverse epigenetic effects of the six major flavonoid subclasses, as well as other natural compounds, in the context of cancer. The goal is to identify new therapeutic epigenetic targets for drug development, whether as stand-alone treatments or in combination with conventional antitumor approaches. Full article
Show Figures

Graphical abstract

17 pages, 2209 KiB  
Article
The Activation of the NF-κB Pathway in Human Adipose-Derived Stem Cells Alters the Deposition of Epigenetic Marks on H3K27 and Is Modulated by Fish Oil
by Jussara de Jesus Simao, Andressa França de Sousa Bispo, Victor Tadeu Gonçalves Plata, Ana Beatriz Marques Abel, Raphael Justa Saran, Júlia Fernandes Barcella, João Carlos Cardoso Alonso, André Valente Santana, Lucia Maria Armelin-Correa and Maria Isabel Cardoso Alonso-Vale
Life 2024, 14(12), 1653; https://doi.org/10.3390/life14121653 - 12 Dec 2024
Cited by 2 | Viewed by 1633
Abstract
Background: Chronic low-grade inflammation in obesity is linked to white adipose tissue (WAT) dysfunction. Plasma lipopolysaccharide (LPS) activates Toll-like receptor 4 (TLR4), triggering NF-κB and worsening these disturbances. Previously, we showed that histone H3 lysine 27 (H3K27) epigenetic modifications affect WAT gene expression [...] Read more.
Background: Chronic low-grade inflammation in obesity is linked to white adipose tissue (WAT) dysfunction. Plasma lipopolysaccharide (LPS) activates Toll-like receptor 4 (TLR4), triggering NF-κB and worsening these disturbances. Previously, we showed that histone H3 lysine 27 (H3K27) epigenetic modifications affect WAT gene expression in high-fat-diet mice, identifying key pathways in adipose-derived stem cells (ASCs). This study explores whether NF-κB influences H3K27 modifiers in human ASCs and evaluates fish oil (FO) as a modulator. Methods: Human visceral WAT ASCs were stimulated with LPS and treated with FO enriched with eicosapentaenoic acid (EPA). Flow cytometry, PCR array, RT-PCR, and Western blot assays were used. Results: LPS increased NF-κB activity, elevating KDM6B demethylase levels and H3K27 acetylation. These epigenetic modifications in LPS-stimulated ASCs were associated with persistent changes in the expression of genes involved in adipogenesis, metabolic regulation, and inflammation, even after LPS removal and cell differentiation. FO mitigated these effects, reducing H3K27 acetylation and promoting methylation. Conclusions: FO demonstrates potential in modulating inflammation-induced epigenetic changes and preserving adipocyte function. Full article
(This article belongs to the Section Physiology and Pathology)
Show Figures

Figure 1

18 pages, 2902 KiB  
Article
KDM4 Regulates the Glycolysis of Hemocytes in the Immune Priming of Eriocheir sinensis
by Xinyu Zhao, Xue Qiao, Simiao Yu, Yuhao Jin, Jixiang Niu, Jie Li, Yingmei Xu, Yuehong Yang, Lingling Wang and Linsheng Song
Int. J. Mol. Sci. 2024, 25(23), 13174; https://doi.org/10.3390/ijms252313174 - 7 Dec 2024
Cited by 2 | Viewed by 1148
Abstract
Immune priming confers a sustained, augmented response of innate immune cells to a secondary challenge, a process that is characteristically reliant on metabolic reprogramming. Recent evidence suggests that histone demethylases play essential roles in the immune priming, while its regulation role in the [...] Read more.
Immune priming confers a sustained, augmented response of innate immune cells to a secondary challenge, a process that is characteristically reliant on metabolic reprogramming. Recent evidence suggests that histone demethylases play essential roles in the immune priming, while its regulation role in the metabolic reprogramming remains largely unknown. In the present study, the concentration of glucose was significantly down-regulated in the hemocytes of crab Eriocheir sinensis after secondary stimulation with Aeromonas hydrophila, while the expression levels of phosphofructokinase (EsPFK) pyruvate kinase (EsPK), hexokinase-2 (EsHK-2) and Glucose-6-phosphate dehydrogenase (EsG-6-PD), along with the concentrations of lactate and the ratio of NAD+/NADH, were elevated. Additionally, the levels of H3K9me3 and its enrichment at the promoters of EsPFK and EsG-6-PD were significantly decreased at 7 days after A. hydrophila stimulation. The lysine Demethylase 4 homologue (EsKDM4) was observed to translocate into the nucleus of crab hemocytes after A. hydrophila stimulation, and its activity markedly increased after secondary stimulation with A. hydrophila. Following RNA interference of EsKDM4, there was a significant increase in H3K9me3 levels, and the enrichment of H3K9me3 at the EsPFK and EsG-6-PD promoters, as well as the concentration of glucose, in the hemocytes of crabs after secondary stimulation with A. hydrophila. Furthermore, mRNA transcripts of EsPFK and EsG-6-PD, as well as the concentration of lactate and ratio of NAD+/NADH, significantly decreased after secondary stimulation. These results suggested that EsKDM4 mediates the enrichment of H3K9me3 at the promoters of EsPFK and EsG-6-PD, thereby regulating glycolysis during the immune priming of crabs. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

25 pages, 3309 KiB  
Review
Epigenetic Modifiers: Exploring the Roles of Histone Methyltransferases and Demethylases in Cancer and Neurodegeneration
by Lauren Reed, Janak Abraham, Shay Patel and Shilpa S. Dhar
Biology 2024, 13(12), 1008; https://doi.org/10.3390/biology13121008 - 3 Dec 2024
Viewed by 2350
Abstract
Histone methyltransferases (HMTs) and histone demethylases (HDMs) are critical enzymes that regulate chromatin dynamics and gene expression through the addition and removal of methyl groups on histone proteins. HMTs, such as PRC2 and SETD2, are involved in the trimethylation of histone H3 at [...] Read more.
Histone methyltransferases (HMTs) and histone demethylases (HDMs) are critical enzymes that regulate chromatin dynamics and gene expression through the addition and removal of methyl groups on histone proteins. HMTs, such as PRC2 and SETD2, are involved in the trimethylation of histone H3 at lysine 27 and lysine 36, influencing gene silencing and activation. Dysregulation of these enzymes often leads to abnormal gene expression and contributes to tumorigenesis. In contrast, HDMs including KDM7A and KDM2A reverse these methylation marks, and their dysfunction can drive disease progression. In cancer, the aberrant activity of specific HMTs and HDMs can lead to the silencing of tumor suppressor genes or the activation of oncogenes, facilitating tumor progression and resistance to therapy. Conversely, in neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and Huntington’s disease (HD), disruptions in histone methylation dynamics are associated with neuronal loss, altered gene expression, and disease progression. We aimed to comprehend the odd activity of HMTs and HDMs and how they contribute to disease pathogenesis, highlighting their potential as therapeutic targets. By advancing our understanding of these epigenetic regulators, this review provides new insights into their roles in cancer and neurodegenerative diseases, offering a foundation for future research. Full article
(This article belongs to the Special Issue Epigenetic Modifications and Changes in Neurodegenerative Diseases)
Show Figures

Figure 1

19 pages, 1339 KiB  
Review
The Tumour Microenvironment and Epigenetic Regulation in BRCA1 Pathogenic Variant-Associated Breast Cancers
by Jun Yu Tay, Josh Xingchong Ho, Fan Foon Cheo and Jabed Iqbal
Cancers 2024, 16(23), 3910; https://doi.org/10.3390/cancers16233910 - 21 Nov 2024
Viewed by 1696
Abstract
Background/Objectives: BRCA1 pathogenic variant (PV)-associated breast cancers are most commonly seen in hereditary genetic conditions such as the autosomal-dominant Hereditary Breast and Ovarian Cancer (HBOC) syndrome, and rarely in sporadic breast cancer. Such breast cancers tend to exhibit greater aggressiveness and poorer [...] Read more.
Background/Objectives: BRCA1 pathogenic variant (PV)-associated breast cancers are most commonly seen in hereditary genetic conditions such as the autosomal-dominant Hereditary Breast and Ovarian Cancer (HBOC) syndrome, and rarely in sporadic breast cancer. Such breast cancers tend to exhibit greater aggressiveness and poorer prognoses due to the influence of BRCA1 pathogenic variants (PVs) on the tumour microenvironment. Additionally, while the genetic basis of BRCA1 PV breast cancer is well-studied, the role of epigenetic mediators in the tumourigenesis of these hereditary breast cancers is also worth exploring. Results: PVs in the BRCA1 gene interact with stromal cells and immune cells, promoting epithelial–mesenchymal transition, angiogenesis, and affecting oestrogen levels. Additionally, BRCA1 PVs contribute to breast cancer development through epigenetic effects on cells, including DNA methylation and histone acetylation, leading to the suppression of proto-oncogenes and dysregulation of cytokines. In terms of epigenetics, lysine-specific demethylase 1 (LSD-1) is considered a master epigenetic regulator, governing both transcriptional repression and activation. It exerts epigenetic control over BRCA1 and, to a lesser extent, BRCA2 genes. The upregulation of LSD-1 is generally associated with a poorer prognosis in cancer patients. In the context of breast cancer in BRCA1/2 PV carriers, LSD-1 contributes to tumour development through various mechanisms. These include the maintenance of a hypoxic environment and direct suppression of BRCA1 gene expression. Conclusions: While LSD-1 itself does not directly cause mutations in BRCA1 or BRCA2 genes, its epigenetic influence sheds light on the potential role of LSD-1 inhibitors as a therapeutic approach in managing breast cancer, particularly in individuals with BRCA1/2 PVs. Targeting LSD-1 may help counteract its detrimental effects and provide a promising avenue for therapy in this specific subgroup of breast cancer. Full article
(This article belongs to the Special Issue Genetics and Epigenetics of Gynecological Cancer)
Show Figures

Figure 1

16 pages, 2486 KiB  
Article
Targeting KDM1A in Neuroblastoma with NCL-1 Induces a Less Aggressive Phenotype and Suppresses Angiogenesis
by Annika Sprüssel, Takayoshi Suzuki, Naoki Miyata, Kathy Astrahantseff, Annabell Szymansky, Joern Toedling, Theresa M. Thole-Kliesch, Annika Ballagee, Marco Lodrini, Annette Künkele, Matthias Truss, Lukas C. Heukamp, Susanne Mathia, Falk Hertwig, Christian Rosenberger, Angelika Eggert, Hedwig E. Deubzer and Johannes H. Schulte
J. Clin. Med. 2024, 13(20), 6081; https://doi.org/10.3390/jcm13206081 - 12 Oct 2024
Viewed by 1506
Abstract
Background: The KDM1A histone demethylase regulates the cellular balance between proliferation and differentiation, and is often deregulated in human cancers including the childhood tumor neuroblastoma. We previously showed that KDM1A is strongly expressed in undifferentiated neuroblastomas and correlates with poor patient prognosis, suggesting [...] Read more.
Background: The KDM1A histone demethylase regulates the cellular balance between proliferation and differentiation, and is often deregulated in human cancers including the childhood tumor neuroblastoma. We previously showed that KDM1A is strongly expressed in undifferentiated neuroblastomas and correlates with poor patient prognosis, suggesting a possible clinical benefit from targeting KDM1A. Methods: Here, we tested the efficacy of NCL-1, a small molecule specifically inhibiting KDM1A in preclinical models for neuroblastoma. Results: NCL-1 mimicked the effects of siRNA-mediated KDM1A knockdown and effectively inhibited KDM1A activity in four neuroblastoma cell lines and a patient-representative cell model. KDM1A inhibition shifted the aggressive tumor cell phenotypes towards less aggressive phenotypes. The proliferation and cell viability was reduced, accompanied by the induction of markers of neuronal differentiation. Interventional NCL-1 treatment of nude mice harboring established neuroblastoma xenograft tumors reduced tumor growth and inhibited cell proliferation. Reduced vessel density and defects in blood vessel construction also resulted, and NCL-1 inhibited the growth and tube formation of HUVEC-C cells in vitro. Conclusions: Inhibiting KDM1A could attack aggressive neuroblastomas two-fold, by re-directing tumor cells toward a less aggressive, slower-growing phenotype and by preventing or reducing the vascular support of large tumors. Full article
(This article belongs to the Special Issue High-Risk Neuroblastoma: New Clinical Insights and Challenges)
Show Figures

Graphical abstract

14 pages, 2060 KiB  
Article
Unraveling the Role of JMJD1B in Genome Stability and the Malignancy of Melanomas
by Perla Cruz, Diego Peña-Lopez, Diego Figueroa, Isidora Riobó, Vincenzo Benedetti, Francisco Saavedra, Claudia Espinoza-Arratia, Thelma M. Escobar, Alvaro Lladser and Alejandra Loyola
Int. J. Mol. Sci. 2024, 25(19), 10689; https://doi.org/10.3390/ijms251910689 - 4 Oct 2024
Cited by 1 | Viewed by 1271
Abstract
Genome instability relies on preserving the chromatin structure, with any histone imbalances threating DNA integrity. Histone synthesis occurs in the cytoplasm, followed by a maturation process before their nuclear translocation. This maturation involves protein folding and the establishment of post-translational modifications. Disruptions in [...] Read more.
Genome instability relies on preserving the chromatin structure, with any histone imbalances threating DNA integrity. Histone synthesis occurs in the cytoplasm, followed by a maturation process before their nuclear translocation. This maturation involves protein folding and the establishment of post-translational modifications. Disruptions in this pathway hinder chromatin assembly and contribute to genome instability. JMJD1B, a histone demethylase, not only regulates gene expression but also ensures a proper supply of histones H3 and H4 for the chromatin assembly. Reduced JMJD1B levels lead to the cytoplasmic accumulation of histones, causing defects in the chromatin assembly and resulting in DNA damage. To investigate the role of JMJD1B in regulating genome stability and the malignancy of melanoma tumors, we used a JMJD1B/KDM3B knockout in B16F10 mouse melanoma cells to perform tumorigenic and genome instability assays. Additionally, we analyzed the transcriptomic data of human cutaneous melanoma tumors. Our results show the enhanced tumorigenic properties of JMJD1B knockout melanoma cells both in vitro and in vivo. The γH2AX staining, Micrococcal Nuclease sensitivity, and comet assays demonstrated increased DNA damage and genome instability. The JMJD1B expression in human melanoma tumors correlates with a lower mutational burden and fewer oncogenic driver mutations. Our findings highlight JMJD1B’s role in maintaining genome integrity by ensuring a proper histone supply to the nucleus, expanding its function beyond gene expression regulation. JMJD1B emerges as a crucial player in preserving genome stability and the development of melanoma, with a potential role as a safeguard against oncogenic mutations. Full article
(This article belongs to the Special Issue Molecular Research on Epigenetic Modifications)
Show Figures

Figure 1

Back to TopTop