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

Journals

Article Types

Countries / Regions

Search Results (156)

Search Parameters:
Keywords = PRMT1

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
26 pages, 2284 KB  
Article
Obesity/Overweight as a Meaningful Modifier of Associations Between Gene Polymorphisms Affecting the Sex Hormone-Binding Globulin Content and Uterine Myoma
by Marina Ponomarenko, Evgeny Reshetnikov, Maria Churnosova, Inna Aristova, Maria Abramova, Vitaly Novakov, Vladimir Churnosov, Alexey Polonikov, Mikhail Churnosov and Irina Ponomarenko
Life 2025, 15(9), 1459; https://doi.org/10.3390/life15091459 - 17 Sep 2025
Viewed by 199
Abstract
The main goal of this study was to consider the role of obesity/overweight as a potential modifier of associations between gene single nucleotide polymorphisms (SNPs) affecting the sex hormone-binding globulin level (SHBGlevel) and uterine myoma (UM). In the two women cohorts [...] Read more.
The main goal of this study was to consider the role of obesity/overweight as a potential modifier of associations between gene single nucleotide polymorphisms (SNPs) affecting the sex hormone-binding globulin level (SHBGlevel) and uterine myoma (UM). In the two women cohorts differentiated by body mass index (BMI) (BMI ≥ 25, n = 782 [379 UM/403 control] and BMI < 25, n = 760 [190 UM/570 control]), the association of genome-wide association studies (GWAS)-correlated SHBGlevel-tied nine loci with UM was studied by method logistic regression with a subsequent in-depth evaluation of the functionality of UM-causal loci and their strongly linked variants. BMI-conditioned differences in the associations of SHBGlevel-tied loci with UM were revealed: in the BMI < 25 group, a variant rs17496332 (A/G) PRMT6 was UM-correlated (OR = 0.70; pperm = 0.024), and in the BMI ≥ 25 cohort, a SNP rs3779195 (T/A) BAIAP2L1 was UM-associated (OR = 1.53; pperm = 0.019). Both the UM-causal loci and their proxy SNPs have pronounced probable functionality in the organism as a whole, as well as in the liver (the SHBG synthesis place), adipose tissue, uterus, etc., thereby influencing significant processes for UM biology such as regulation of the gene transcription, embryogenesis/development, cell proliferation/differentiation/apoptosis, metabolism, lipid exchange, etc. In conclusion, the results of our work demonstrated, for the first time, the essential role of obesity/overweight as a meaningful modifier of associations between SHBGlevel-tied polymorphisms and UM. Full article
Show Figures

Figure 1

26 pages, 644 KB  
Review
Therapeutic Targeting of Protein Lysine and Arginine Methyltransferases: Principles and Strategies for Inhibitor Design
by Isaac Micallef and Byron Baron
Int. J. Mol. Sci. 2025, 26(18), 9038; https://doi.org/10.3390/ijms26189038 - 17 Sep 2025
Cited by 1 | Viewed by 432
Abstract
Standard cancer chemotherapy is increasingly being supplemented with novel therapeutics to overcome known chemoresistance pathways. Resistance to treatment is common across various tumour types, driven by multiple mechanisms. One emerging contributor is protein methylation, a post-translational modification mediated by protein methyltransferases (PMTs), which [...] Read more.
Standard cancer chemotherapy is increasingly being supplemented with novel therapeutics to overcome known chemoresistance pathways. Resistance to treatment is common across various tumour types, driven by multiple mechanisms. One emerging contributor is protein methylation, a post-translational modification mediated by protein methyltransferases (PMTs), which regulate protein function by adding methyl groups, mainly on lysine and arginine residues. Dysregulation of protein lysine methyltransferases (PKMTs) and protein arginine methyltransferases (PRMTs) has been linked to cancer progression and drug resistance, making them attractive therapeutic targets. Consequently, several small-molecule PMT inhibitors have been developed, with some progressing to clinical trials. However, many candidates showing promise in preclinical studies fail to demonstrate efficacy or safety in later stages, limiting clinical success. This gap highlights the need to rethink current approaches to PMT inhibitor design. A deeper understanding of PMT mechanisms, catalytic domains, and their roles in chemoresistance is essential for creating more selective, potent, and clinically viable inhibitors. This review will summarise major chemoresistance pathways and PMTs implicated in cancer, then explore current and prospective PMT inhibitor classes. Building on mechanistic insights, we propose strategies to develop next-generation inhibitors with improved therapeutic potential against chemoresistant cancers. Full article
(This article belongs to the Special Issue Protein Methyltransferases in Human Health and Diseases)
Show Figures

Figure 1

24 pages, 4860 KB  
Review
Targeting PRMT5: Current Inhibitors and Emerging Strategies for Therapeutic Intervention
by Zhihang Shen and Chenglong Li
Processes 2025, 13(9), 2878; https://doi.org/10.3390/pr13092878 - 9 Sep 2025
Viewed by 748
Abstract
Epigenetic dysregulation is a hallmark of tumorigenesis, with arginine methylation—a post-translational modification—emerging as a key regulatory mechanism in cancer biology. This modification, catalyzed by protein arginine methyltransferases (PRMTs), influences critical cellular processes, including proliferation, differentiation, transcription, RNA splicing, DNA repair, and immune signaling. [...] Read more.
Epigenetic dysregulation is a hallmark of tumorigenesis, with arginine methylation—a post-translational modification—emerging as a key regulatory mechanism in cancer biology. This modification, catalyzed by protein arginine methyltransferases (PRMTs), influences critical cellular processes, including proliferation, differentiation, transcription, RNA splicing, DNA repair, and immune signaling. Among the PRMT family, PRMT5 has garnered significant attention due to its elevated expression across various solid tumors and hematological malignancies, and its strong association with poor clinical outcomes. Notably, PRMT5 exhibits a unique vulnerability in methyl-thio-adenosine phosphorylase (MTAP)-deficient cancers, making it an attractive therapeutic target. Recent advances have led to the development of several PRMT5 inhibitors with diverse binding modes, some of which have progressed into clinical trials for advanced cancers. This review provides a structural and mechanistic overview of PRMT5, summarizes current inhibition strategies, and discusses the challenges and future directions in targeting PRMT5 for cancer therapy. Full article
(This article belongs to the Special Issue Pharmaceutical Development and Bioavailability Analysis, 2nd Edition)
Show Figures

Graphical abstract

9 pages, 1106 KB  
Communication
PRMT5 Inhibition as a Potential Strategy for KRAS Mutant CRC: Downstream Mediators of the PRMT5–KRAS Crosstalk
by Mark Spivak, Moshe Pahmer, Dorna Delrahimnia, Tzuriel Sapir and David Shifteh
Curr. Issues Mol. Biol. 2025, 47(8), 665; https://doi.org/10.3390/cimb47080665 - 18 Aug 2025
Viewed by 546
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide with KRAS mutations present in nearly 45% of cases. Compared to KRAS wild-type (WT) CRC, KRAS-mutant CRC is associated with poorer prognosis and fewer effective treatment options. Protein Arginine Methyltransferase 5 (PRMT5), [...] Read more.
Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide with KRAS mutations present in nearly 45% of cases. Compared to KRAS wild-type (WT) CRC, KRAS-mutant CRC is associated with poorer prognosis and fewer effective treatment options. Protein Arginine Methyltransferase 5 (PRMT5), an epigenetic regulator involved in diverse cellular processes, is currently under investigation as a therapeutic target in multiple cancer types. Our previous work demonstrated that PRMT5 inhibition produces stronger therapeutic effects in KRAS-mutant CRC cells than in KRAS WT cells, suggesting potential crosstalk between PRMT5 and KRAS. In this study, we aimed to identify key downstream proteins that may mediate this interaction. Through a literature review, protein–protein interaction analysis (STRING database), gene expression analysis (GEPIA database), and correlation analysis (GEPIA database), we identified MYC, E2F1, and EIF4E as critical candidates. These proteins are shown to interact with both PRMT5 and KRAS in STRING, are overexpressed in CRC tumor samples, and show positive gene expression correlations with PRMT5 and KRAS in patient data. These findings are significant, as they provide new insights into the PRMT5–KRAS crosstalk and suggest potential targets for novel and combination therapies in KRAS-mutant CRC. Further research and biological experiments are needed to verify and outline the exact molecular processes behind MYC, E2F1, and EIF4E’s interactions with both PRMT5 and KRAS. Full article
(This article belongs to the Section Bioinformatics and Systems Biology)
Show Figures

Figure 1

16 pages, 283 KB  
Review
Current and Emerging Therapies for Targeting Protein Arginine Methyltransferases (PRMTs) in Cancer
by Adriana Kaganovski, Bayle Smith-Salzberg, Hadar K. Shimshon, Andrew Draheim, Mark Spivak, Tzuriel Sapir and David Shifteh
Int. J. Mol. Sci. 2025, 26(16), 7907; https://doi.org/10.3390/ijms26167907 - 16 Aug 2025
Viewed by 727
Abstract
Protein arginine methyltransferases (PRMTs) are a class of enzymes that mediate critical post-translational modifications through arginine methylation as epigenetic regulators. PRMTs have been shown to have a vast array of regulatory effects including in gene expression, signal transduction, and cellular proliferation. Dysregulation of [...] Read more.
Protein arginine methyltransferases (PRMTs) are a class of enzymes that mediate critical post-translational modifications through arginine methylation as epigenetic regulators. PRMTs have been shown to have a vast array of regulatory effects including in gene expression, signal transduction, and cellular proliferation. Dysregulation of PRMT activity has been seen in the progression of various cancers, including breast, lung, and colorectal cancer. Moreover, PRMT overexpression has been shown to correlate with poor patient prognosis. This review aims to explore the roles of the individual PRMTs in cancer and aims to highlight the latest and newest developments of PRMT inhibitors as emerging therapeutic strategies. Numerous preclinical and clinical studies have identified several novel compounds that effectively target PRMT activity and have shown significant therapeutic results. As such, this review aims to not only highlight the current research findings, but to also emphasize the significant need for future research on PRMTs as novel therapeutic targets in cancer. Full article
(This article belongs to the Section Molecular Oncology)
20 pages, 1516 KB  
Review
Ferroptosis and Nrf2 Signaling in Head and Neck Cancer: Resistance Mechanisms and Therapeutic Prospects
by Jaewang Lee, Youngin Seo and Jong-Lyel Roh
Antioxidants 2025, 14(8), 993; https://doi.org/10.3390/antiox14080993 - 13 Aug 2025
Cited by 1 | Viewed by 1156
Abstract
Ferroptosis is an iron-dependent form of regulated cell death marked by lipid peroxidation in polyunsaturated phospholipids. In head and neck cancer (HNC), where resistance to chemotherapy and immunotherapy is common, ferroptosis offers a mechanistically distinct strategy to overcome therapeutic failure. However, cancer cells [...] Read more.
Ferroptosis is an iron-dependent form of regulated cell death marked by lipid peroxidation in polyunsaturated phospholipids. In head and neck cancer (HNC), where resistance to chemotherapy and immunotherapy is common, ferroptosis offers a mechanistically distinct strategy to overcome therapeutic failure. However, cancer cells often evade ferroptosis via activation of nuclear factor erythroid 2-related factor 2 (Nrf2), a key regulator of antioxidant and iron-regulatory genes. HNC remains therapeutically challenging due to therapy resistance driven by redox adaptation. This review highlights the ferroptosis pathway—a form of regulated necrosis driven by iron and lipid peroxidation—and its regulation by Nrf2, a master antioxidant transcription factor. We detail how Nrf2 contributes to ferroptosis evasion in HNC and summarize emerging preclinical studies targeting this axis. The review aims to synthesize molecular insights and propose therapeutic perspectives for overcoming resistance in HNC by modulating Nrf2–ferroptosis signaling. We conducted a structured narrative review of the literature using PubMed databases. Relevant studies from 2015 to 2025 focusing on ferroptosis, Nrf2 signaling, and head and neck cancer were selected based on their experimental design, novelty, and relevance to clinical resistance mechanisms. In HNC, Nrf2 mediates resistance through transcriptional upregulation of GPX4 and SLC7A11, epigenetic stabilization by PRMT4 and ALKBH5, and activation by FGF5 and platelet-derived extracellular vesicles. Epstein–Barr virus (EBV) infection also enhances Nrf2 signaling in nasopharyngeal carcinoma. More recently, loss-of-function KEAP1 mutations have been linked to persistent Nrf2 activation and upregulation of NQO1, which confer resistance to both ferroptosis and immune checkpoint therapy. Targeting NQO1 in KEAP1-deficient models restores ferroptosis and reactivates antitumor immunity. Additionally, the natural alkaloid trigonelline has shown promise in reversing Nrf2-mediated ferroptosis resistance in cisplatin-refractory tumors. Pharmacologic agents such as auranofin, fucoxanthin, carnosic acid, and disulfiram/copper complexes have demonstrated efficacy in sensitizing HNC to ferroptosis by disrupting the Nrf2 axis. This review summarizes emerging mechanisms of ferroptosis evasion and highlights therapeutic strategies targeting the Nrf2–ferroptosis network. Integrating ferroptosis inducers with immune and chemotherapeutic approaches may provide new opportunities for overcoming resistance in head and neck malignancies. Full article
(This article belongs to the Special Issue Oxidative Stress and NRF2 in Health and Disease—2nd Edition)
Show Figures

Graphical abstract

16 pages, 2487 KB  
Article
Overexpression of Circular PRMT1 Transcripts in Colorectal Adenocarcinoma Predicts Recurrence and Poor Overall Survival
by Panagiotis Kokoropoulos, Spyridon Christodoulou, Panagiotis Tsiakanikas, Panteleimon Vassiliu, Christos K. Kontos and Nikolaos Arkadopoulos
Int. J. Mol. Sci. 2025, 26(14), 6683; https://doi.org/10.3390/ijms26146683 - 11 Jul 2025
Viewed by 455
Abstract
Colorectal cancer (CRC) is one of the most prevalent and deadly neoplasms globally; this fact puts emphasis on the need for accurate molecular biomarkers for early detection and accurate prognosis. Circular RNAs (circRNAs) have recently emerged as very promising cancer biomarkers. In this [...] Read more.
Colorectal cancer (CRC) is one of the most prevalent and deadly neoplasms globally; this fact puts emphasis on the need for accurate molecular biomarkers for early detection and accurate prognosis. Circular RNAs (circRNAs) have recently emerged as very promising cancer biomarkers. In this study, we thoroughly examined whether the expression levels of circular transcripts of the protein arginine methyltransferase 1 (PRMT1) gene can predict the prognosis of patients diagnosed with colorectal adenocarcinoma, the most frequent type of CRC. Hence, a highly sensitive quantitative PCR (qPCR) assay was developed and applied to quantify circ-PRMT1 expression in cDNAs from 210 primary colorectal adenocarcinoma tissue specimens and 86 paired normal colorectal mucosae. Extensive biostatistical analysis was then performed to assess the potential prognostic power of circ-PRMT1. Significant overexpression of this molecule was observed in colorectal adenocarcinoma tissue samples in contrast to their non-cancerous counterparts. Moreover, higher circ-PRMT1 expression was correlated with poorer disease-free survival (DFS) and worse overall survival (OS) in colorectal adenocarcinoma patients. Interestingly, multivariate Cox regression analysis revealed that the prognostic value of the expression of this circRNA does not depend on other established prognostic factors included in the prognostic model. Furthermore, the stratification of patients based on TNM staging revealed that higher circ-PRMT1 levels were significantly related to shorter DFS and OS intervals, particularly in patients with colorectal adenocarcinoma of TNM stage II or III. In summary, this original research study provides evidence that circ-PRMT1 overexpression represents a promising molecular biomarker of poor prognosis in colorectal adenocarcinoma, not depending on other established prognostic factors such as TNM staging. Full article
(This article belongs to the Special Issue New Molecular Aspects of Colorectal Cancer)
Show Figures

Figure 1

24 pages, 14721 KB  
Article
Loss of 4.1B Drives PRMT3-Mediated Regulation of GBM Brain Tumour Stem Cell Growth
by Ravinder K. Bahia, Kyle Heemskerk, Samir Assaf, Orsolya Cseh, Xiaoguang Hao, Rozina Hassam, Panagiotis Prinos, H. Artee Luchman and Samuel Weiss
Int. J. Transl. Med. 2025, 5(3), 29; https://doi.org/10.3390/ijtm5030029 - 7 Jul 2025
Viewed by 760
Abstract
Background: Protein arginine methyltransferase 3 (PRMT3), a type I family PRMT, regulates the activity of downstream substrates by catalyzing the asymmetric dimethylation of arginine residues. While PRMT3 activity has been reported to be deregulated in many cancers, including glioblastoma (GBM), the underlying signalling [...] Read more.
Background: Protein arginine methyltransferase 3 (PRMT3), a type I family PRMT, regulates the activity of downstream substrates by catalyzing the asymmetric dimethylation of arginine residues. While PRMT3 activity has been reported to be deregulated in many cancers, including glioblastoma (GBM), the underlying signalling mechanisms that contribute to disease progression are largely unknown. Methods: We tested the efficacy of a PRMT3 chemical probe, SGC707, in a cohort of GBM patient-derived primary and recurrent brain tumour stem cell (BTSC) lines. RNA-sequencing, CRISPR-cas9 knockout, and inducible overexpression methods were used to investigate the molecular mechanisms regulated by the aberrant activity of PRMT3 in different BTSC lines. Results: We show that expression of the tumour suppressor protein 4.1B, a negative regulator of PRMT3, predicts the response of GBM BTSCs to the PRMT3 chemical probe, SGC707. Furthermore, PRMT3 modulates the stability and subcellular localization of the downstream effector, UHRF1, a member of the DNA methylation complex. These findings suggest that UHRF1 and DNMT1 may suppress the expression of 4.1B through the increased promoter methylation of EPB4.1L3. Intriguingly, the inducible overexpression of EPB4.1L3 in the BT248EPB4.1L3low BTSC line mimicked the effects of the pharmacologic and genetic inhibition of PRMT3. In contrast, knockout of EPB4.1L3 in BT143EPB4.1L3high cells reduced the interactions between PRMT3 and 4.1B proteins, resulting in increased sensitivity of knockout cells to SGC707 treatment. Conclusions: These findings show that 4.1B, PRMT3, and UHRF1/DNMT1 function together to promote BTSC growth. Thus, targeting PRMT3 or UHRF1/DNMT1, especially in tumours with low endogenous 4.1B protein, may have high therapeutic relevance. Full article
Show Figures

Figure 1

15 pages, 4150 KB  
Article
PRMT5 Identified as a Viable Target for Combination Therapy in Preclinical Models of Pancreatic Cancer
by Xiaolong Wei, William J. Kane, Sara J. Adair, Sarbajeet Nagdas, Denis Liu and Todd W. Bauer
Biomolecules 2025, 15(7), 948; https://doi.org/10.3390/biom15070948 - 30 Jun 2025
Viewed by 801
Abstract
Pancreatic cancer is the third leading cause of cancer-related death in the US. First-line chemotherapy regimens for pancreatic ductal adenocarcinoma (PDAC) include FOLFIRINOX or gemcitabine (Gem) with or without paclitaxel (Ptx); however, 5-year survival with these regimens remains poor. Previous work has demonstrated [...] Read more.
Pancreatic cancer is the third leading cause of cancer-related death in the US. First-line chemotherapy regimens for pancreatic ductal adenocarcinoma (PDAC) include FOLFIRINOX or gemcitabine (Gem) with or without paclitaxel (Ptx); however, 5-year survival with these regimens remains poor. Previous work has demonstrated protein arginine methyltransferase 5 (PRMT5) to be a promising therapeutic target in combination with Gem for the treatment of PDAC; however, these findings have yet to be confirmed in relevant preclinical models of PDAC. To test the possibility of PRMT5 as a viable therapeutic target, clinically relevant orthotopic and metastatic patient-derived xenograft (PDX) mouse models of PDAC growth were utilized to evaluate the effect of PRMT5 knockout (KO) or pharmacologic inhibition on treatment with Gem alone or Gem with Ptx. Primary endpoints included tumor volume, tumor weight, or metastatic tumor burden as appropriate. The results showed that Gem-treated PRMT5 KO tumors exhibited decreased growth and were smaller in size compared to Gem-treated wild-type (WT) tumors. Similarly, the Gem-treated PRMT5 KO metastatic burden was lower than the Gem-treated WT metastatic burden. The addition of a PRMT5 pharmacologic inhibitor to Gem and Ptx therapy resulted in a lower final tumor weight and fewer metastatic tumors. The depletion of PRMT5 results in increased DNA damage in response to Gem and Ptx treatment. Thus, PRMT5 genetic depletion or inhibition in combination with Gem-based therapy improved the response in primary and metastatic PDAC in clinically relevant mouse models, suggesting that PRMT5 is a viable therapeutic target for combination therapy in PDAC. Full article
Show Figures

Figure 1

17 pages, 791 KB  
Review
Exploiting Synthetic Lethality of PRMT5 for Precision Treatment of MTAP-Deficient Glioblastoma
by Trang T. T. Nguyen, Eunhee Yi and Christian E. Badr
Int. J. Transl. Med. 2025, 5(3), 27; https://doi.org/10.3390/ijtm5030027 - 29 Jun 2025
Viewed by 1997
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, characterized by a dismal prognosis and limited therapeutic options. Its highly invasive nature and pronounced intratumoral heterogeneity underscores the urgent need for innovative and targeted therapeutic strategies. One promising approach is synthetic [...] Read more.
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, characterized by a dismal prognosis and limited therapeutic options. Its highly invasive nature and pronounced intratumoral heterogeneity underscores the urgent need for innovative and targeted therapeutic strategies. One promising approach is synthetic lethality, which exploits cancer-specific genetic vulnerabilities to selectively eliminate tumor cells. A well-characterized example involves the deletion of methylthioadenosine phosphorylase (MTAP), commonly observed in GBM and other malignancies. This review focuses on synthetic lethality targeting protein arginine methyltransferase 5 (PRMT5) in MTAP-deleted GBM. Loss of MTAP leads to the accumulation of methylthioadenosine (MTA), a metabolite that partially inhibits PRMT5, thereby creating a selective vulnerability to PRMT5 inhibition which is used to inhibit the residual function of PRMT5. We critically evaluate preclinical and clinical data on both first- and second-generation PRMT5 inhibitors, with particular emphasis on MTA-cooperative compounds that selectively exploit MTAP deficiency. Despite promising anti-tumor activity in vitro, the clinical efficacy of PRMT5 inhibitors is often limited by the tumor microenvironment, particularly the impact of non-malignant cells that attenuate drug activity. Finally, we explore rational combination strategies that integrate PRMT5 inhibition with existing therapies to enhance clinical outcomes in GBM. Full article
Show Figures

Figure 1

19 pages, 1157 KB  
Article
Examination of Genetic and Epigenetic Characteristics of Patients with Hyperhomocysteinemia Following High-Dose Folic Acid Consumption
by Barbara K. Bartak, Zsofia B. Nagy, Nikolett Szakallas, Alexandra Kalmar, Eszter Farkas, Fruzsina Banyai, Orsolya Pipek, Istvan Csabai, Nora Sydo, Emese Csulak, Bela Merkely, Istvan Takacs and Bela Molnar
Nutrients 2025, 17(13), 2133; https://doi.org/10.3390/nu17132133 - 27 Jun 2025
Viewed by 788
Abstract
Purpose: Homocysteine (HCY) metabolism is regulated by the methionine cycle, which is essential for DNA methylation and is associated with the folate cycle. This study examines the alterations in DNA methylation signature including epigenetic age changes, measure cell-free DNA (cfDNA), and HCY concentrations, [...] Read more.
Purpose: Homocysteine (HCY) metabolism is regulated by the methionine cycle, which is essential for DNA methylation and is associated with the folate cycle. This study examines the alterations in DNA methylation signature including epigenetic age changes, measure cell-free DNA (cfDNA), and HCY concentrations, and identifies genetic markers that may influence homocysteine response following folic acid (FA) supplementation in individuals with hyperhomocysteinemia (HHC). Methods: Blood samples were obtained from 43 HHC patients undergoing FA supplementation. We quantified FA and HCY levels, separated plasma and white blood cell fractions, and evaluated global DNA methylation using LINE-1 bisulfite pyrosequencing. Biological age was determined using Illumina BeadArray technology, and whole-exome sequencing was performed to investigate the patients’ genetic backgrounds. Results: Following FA supplementation, cfDNA levels significantly decreased and correlated positively with HCY (r = 0.2375). Elevated average LINE-1 methylation of cfDNA and PBMC-origin DNA was observed, with mean relative changes of 1.9% for both sample types. Regarding HCY levels, we categorized patients based on their response to FA supplementation. FA responders showed decreased HCY from 15.7 ± 5.5 to 11 ± 2.9 µmol/L, while in FA non-responders, an opposite trend was detected. The average biological age was reduced by 2.6 years, with a notable reduction observed in 80% of non-responders and 48% of responders. Sequencing identified mutations in several genes related to the one-carbon cycle, including MTRR, CHAT, and MTHFD1, with strong correlations to the non-responder phenotypes found in genes like PRMT3, TYMS, DNMT3A, and HIF3A. Conclusions: FA supplementation influences the HCY level, as well as affects the cfDNA amount and the DNA methylation pattern. However, genetic factors may play a crucial role in mediating individual responses to folate intake, emphasizing the need for personalized approaches in managing hyperhomocysteinemia. Full article
Show Figures

Figure 1

16 pages, 2006 KB  
Review
Functional Dynamics of Arginine Mono- and Di-Methylation
by Xi’ang Wang, Bin Zhu, Robert Winn, Shanfa Lu and Hengbin Wang
Cells 2025, 14(11), 796; https://doi.org/10.3390/cells14110796 - 29 May 2025
Viewed by 896
Abstract
Arginine methylation is a crucial post-translational modification (PTM) that plays a significant role in various biological processes. It occurs in two primary forms: mono-methylation (MMA) and di-methylation (DMA), with the latter further classified into symmetric (SDMA) and asymmetric methylation (ADMA). This review examines [...] Read more.
Arginine methylation is a crucial post-translational modification (PTM) that plays a significant role in various biological processes. It occurs in two primary forms: mono-methylation (MMA) and di-methylation (DMA), with the latter further classified into symmetric (SDMA) and asymmetric methylation (ADMA). This review examines the functional implications of these methylation states, current detection methodologies, proteomics-based analytical approaches, and the different impacts of these methylations on protein function. Finally, the role of protein arginine methyltransferases (PRMTs) and their substrate specificity in shaping the arginine methylome are discussed. Full article
Show Figures

Graphical abstract

17 pages, 1590 KB  
Review
Molecular Mechanisms of Tumor Progression and Novel Therapeutic and Diagnostic Strategies in Mesothelioma
by Taketo Kato, Ichidai Tanaka, Heng Huang, Shoji Okado, Yoshito Imamura, Yuji Nomata, Hirofumi Takenaka, Hiroki Watanabe, Yuta Kawasumi, Keita Nakanishi, Yuka Kadomatsu, Harushi Ueno, Shota Nakamura, Tetsuya Mizuno and Toyofumi Fengshi Chen-Yoshikawa
Int. J. Mol. Sci. 2025, 26(9), 4299; https://doi.org/10.3390/ijms26094299 - 1 May 2025
Cited by 1 | Viewed by 1830
Abstract
Mesothelioma is characterized by the inactivation of tumor suppressor genes, with frequent mutations in neurofibromin 2 (NF2), BRCA1-associated protein 1 (BAP1), and cyclin-dependent kinase inhibitor 2A (CDKN2A). These mutations lead to disruptions in the Hippo signaling pathway [...] Read more.
Mesothelioma is characterized by the inactivation of tumor suppressor genes, with frequent mutations in neurofibromin 2 (NF2), BRCA1-associated protein 1 (BAP1), and cyclin-dependent kinase inhibitor 2A (CDKN2A). These mutations lead to disruptions in the Hippo signaling pathway and histone methylation, thereby promoting tumor growth. NF2 mutations result in Merlin deficiency, leading to uncontrolled cell proliferation, whereas BAP1 mutations impair chromatin remodeling and hinder DNA damage repair. Emerging molecular targets in mesothelioma include mesothelin (MSLN), oxytocin receptor (OXTR), protein arginine methyltransferase (PRMT5), and carbohydrate sulfotransferase 4 (CHST4). MSLN-based therapies, such as antibody–drug conjugates and immunotoxins, have shown efficacy in clinical trials. OXTR, upregulated in mesothelioma, is correlated with poor prognosis and represents a novel therapeutic target. PRMT5 inhibition is being explored in tumors with MTAP deletions, commonly co-occurring with CDKN2A loss. CHST4 expression is associated with improved prognosis, potentially influencing tumor immunity. Immune checkpoint inhibitors targeting PD-1/PD-L1 have shown promise in some cases; however, resistance mechanisms remain a challenge. Advances in multi-omics approaches have improved our understanding of mesothelioma pathogenesis. Future research will aim to identify novel therapeutic targets and personalized treatment strategies, particularly in the context of epigenetic therapy and combination immunotherapy. Full article
(This article belongs to the Special Issue Translational Oncology: From Molecular Basis to Therapy)
Show Figures

Figure 1

19 pages, 13715 KB  
Article
PRMT7-Mediated PTEN Activation Enhances Bone Regeneration in Female Mice
by Yingfei Zhang, Jia Qing, Yang Li, Xin Gao, Dazhuang Lu, Yiyang Wang, Lanxin Gu, Hui Zhang, Zechuan Li, Xu Wang, Yongsheng Zhou and Ping Zhang
Int. J. Mol. Sci. 2025, 26(7), 2981; https://doi.org/10.3390/ijms26072981 - 25 Mar 2025
Viewed by 532
Abstract
Epigenetic regulation provides new insights into the mechanisms of osteogenic differentiation and identifies potential targets for treating bone-related diseases. However, the specific regulatory networks and mechanisms involved still need further investigation. In this study, we identify PRMT7 as a novel epigenetic regulator of [...] Read more.
Epigenetic regulation provides new insights into the mechanisms of osteogenic differentiation and identifies potential targets for treating bone-related diseases. However, the specific regulatory networks and mechanisms involved still need further investigation. In this study, we identify PRMT7 as a novel epigenetic regulator of mesenchymal stem cells (MSCs) osteogenic commitment. Conditional knockout of Prmt7 in mice reveals a significant impairment in osteogenesis and bone regeneration, specifically in females, affecting both femurs and mandibles, with no noticeable effect in males. Mechanistically, PRMT7 modulates MSCs osteogenic differentiation by activating PTEN. Specifically, PRMT7 enhances PTEN transcription by increasing H3R2me1 levels at the PTEN promoter. Additionally, PRMT7 interacts with the PTEN protein and stabilizes nuclear PTEN, revealing an unprecedented pathway. Notably, overexpression of PTEN alleviates the osteogenic deficits observed in Prmt7-deficient mice. This research establishes PRMT7 as a potential therapeutic target for promoting bone formation/regeneration and offers novel molecular insights into the PRMT7–PTEN regulatory axis, underscoring its significance in bone biology and regenerative medicine. Full article
(This article belongs to the Special Issue Cells and Molecules in Bone Remodeling and Repair)
Show Figures

Figure 1

12 pages, 3475 KB  
Article
Alternative Splicing Analysis Reveals Adrenergic Signaling as a Novel Target for Protein Arginine Methyltransferase 5 (PRMT5) in the Heart
by Shouye Jiao, Yimeng Zhang, Xiao Yang, Jian Wang and Zhenhua Li
Int. J. Mol. Sci. 2025, 26(5), 2301; https://doi.org/10.3390/ijms26052301 - 5 Mar 2025
Viewed by 913
Abstract
Adrenergic signaling is critical for maintaining cardiac function and works by regulating heart rate, contractility, and stress responses. Protein arginine methyltransferase 5 (PRMT5), a key enzyme involved in gene expression, signal transduction, and RNA processing, has been revealed to be an important factor [...] Read more.
Adrenergic signaling is critical for maintaining cardiac function and works by regulating heart rate, contractility, and stress responses. Protein arginine methyltransferase 5 (PRMT5), a key enzyme involved in gene expression, signal transduction, and RNA processing, has been revealed to be an important factor in heart disease. However, its specific effects on adrenergic signaling have not been fully elucidated. In this study, we examined the role of PRMT5 in the heart by analyzing alternative splicing events in cardiac tissues from Prmt5-deficient mice. High-throughput RNA sequencing and bioinformatics analyses identified significant alterations in alternative splicing, particularly in genes related to adrenergic signaling, which were further validated using reverse transcription PCR. These results underscore the role of PRMT5 as an important regulator of alternative splicing in the heart and identify adrenergic signaling as a novel target. Collectively, our findings offer new insights into the molecular mechanisms underlying cardiac function and suggest that PRMT5 is a potential therapeutic target for heart diseases. Full article
(This article belongs to the Collection Feature Papers in Molecular Genetics and Genomics)
Show Figures

Graphical abstract

Back to TopTop