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 (150)

Search Parameters:
Keywords = PRMT7

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 2487 KiB  
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 220
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 KiB  
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 420
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 KiB  
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 411
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 KiB  
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 974
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 KiB  
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 388
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 KiB  
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 594
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 KiB  
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 1163
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 KiB  
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 382
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 KiB  
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 739
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

22 pages, 1212 KiB  
Review
Protein Arginine Methyltransferase 1: A Multi-Purpose Player in the Development of Cancer and Metabolic Disease
by Daphne de Korte and Menno Hoekstra
Biomolecules 2025, 15(2), 185; https://doi.org/10.3390/biom15020185 - 27 Jan 2025
Viewed by 1515
Abstract
Protein arginine methyltransferase 1 (PRMT1) is the main PRMT family member involved in the formation of monomethylarginine and asymmetric dimethylarginine on its protein substrates. Many protein substrates of PRMT1 are key mediators of cell proliferation and oncogenesis. As such, the function of PRMT1 [...] Read more.
Protein arginine methyltransferase 1 (PRMT1) is the main PRMT family member involved in the formation of monomethylarginine and asymmetric dimethylarginine on its protein substrates. Many protein substrates of PRMT1 are key mediators of cell proliferation and oncogenesis. As such, the function of PRMT1 has been most prominently investigated in the context of cancer development. However, recent in vitro and in vivo studies have highlighted that PRMT1 may also promote metabolic disorders. With the current review, we aim to present an in-depth overview of how PRMT1 influences epigenetic modulation, transcriptional regulation, DNA damage repair, and signal transduction in cancer. Furthermore, we summarize the current knowledge regarding the role of PRMT1 in metabolic reprogramming, lipid metabolism, and glucose metabolism and describe the association of PRMT1 with numerous metabolic pathologies such as obesity, liver disease, and type 2 diabetes. It has become apparent that inhibiting the function of PRMT1 will likely serve as the most beneficial therapeutic approach, since several PRMT1 inhibitors have already been shown to exert positive effects on both cancer and metabolic disease in preclinical settings. However, pharmacological PRMT1 inhibition has not yet been shown to be therapeutically effective in clinical studies. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

12 pages, 1669 KiB  
Article
Histone Arginine Methylation in the Kidneys of Rana sylvatica During Freeze–Thaw Cycle
by Olawale O. Taiwo and Kenneth B. Storey
Kinases Phosphatases 2025, 3(1), 1; https://doi.org/10.3390/kinasesphosphatases3010001 - 7 Jan 2025
Viewed by 1231
Abstract
Freeze tolerance is a remarkable adaptive trait exhibited by wood frogs (Rana sylvatica) during their hibernation period. To show the epigenetic mechanisms that contribute to kidney protection during freezing stress, this present study provides the first investigation of the role and [...] Read more.
Freeze tolerance is a remarkable adaptive trait exhibited by wood frogs (Rana sylvatica) during their hibernation period. To show the epigenetic mechanisms that contribute to kidney protection during freezing stress, this present study provides the first investigation of the role and dynamics of histone arginine methylation and the expression of protein arginine methyltransferases (PRMTs) in a freeze-tolerant vertebrate. Kidney samples from three groups were assessed: (a) control frogs acclimated at 5 °C, (b) 24 h frozen frogs, and (c) 8 h thawed frogs. Our findings revealed significant downregulation of PRMT1, PRMT3, and PRMT5 in kidneys from frozen wood frogs compared to the control group. This downregulation indicates a potential role for PRMT enzymes in the regulation of arginine methylation under freezing stress. In addition, we observed distinct changes in histone marks. H3R17me2a showed significant upregulation after 24 h of freezing, potentially indicating its involvement in the activation of genes related to freezing survival. By contrast, H3R26me2a was downregulated after both 24 h freezing and 8 h thawing, whereas H3R8me2a showed sustained levels after freezing but was downregulated after thawing. These findings highlight the dynamic nature of histone arginine methylation and PRMT expression in wood frog kidneys during freezing–thawing. Our results indicate that epigenetic modifications play a crucial role in shaping the adaptive responses of wood frog kidneys to freezing stress and contribute new information on the underlying biochemical modifications that support vertebrate freeze tolerance. Full article
Show Figures

Figure 1

14 pages, 4233 KiB  
Article
PRMT5 Inhibitor EPZ015666 Decreases the Viability and Encystment of Entamoeba invadens
by Rigoberto Ortiz-Hernández, Elmer Joel Millán-Casarrubias, Jeni Bolaños, Susana Munguía-Robledo, Carlos Vázquez-Calzada, Elisa Azuara-Licéaga, Jesús Valdés and Mario Alberto Rodríguez
Molecules 2025, 30(1), 62; https://doi.org/10.3390/molecules30010062 - 27 Dec 2024
Cited by 1 | Viewed by 1087
Abstract
Protein arginine methyltransferase 5 (PRMT5) is an enzyme that produces monomethyl arginine (MMA) and symmetric dimethyl arginine (sDMA), post-translational modifications that regulate several cellular processes, including stage conversion in parasitic protozoans. Entamoeba histolytica, the etiologic agent of human amebiasis, has two stages [...] Read more.
Protein arginine methyltransferase 5 (PRMT5) is an enzyme that produces monomethyl arginine (MMA) and symmetric dimethyl arginine (sDMA), post-translational modifications that regulate several cellular processes, including stage conversion in parasitic protozoans. Entamoeba histolytica, the etiologic agent of human amebiasis, has two stages in its life cycle, the trophozoite, which is the replicative form, and the cyst, corresponding to the infective phase. The study of the molecular mechanisms that regulate differentiation in this parasite has been overdue because of a lack of efficient protocols for in vitro encystment. For this reason, Entamoeba invadens, a parasite of reptiles, has been used as a differentiation model system for the genus. Here, we demonstrated the presence of sDMA in E. invadens, which increases during encystment, and identified the PRMT5 of this microorganism (EiPRMT5). In addition, we performed 3D modeling of this enzyme, as well as its molecular docking with the PRMT5 inhibitor EPZ015666, which predicted the affinity of the drug for the active site of the enzyme. In agreement with these findings, EPZ015666 reduced trophozoite viability and encystment. Therefore, EiPRMT5 is a potential target for inhibiting the spread of amebiasis. Full article
Show Figures

Graphical abstract

11 pages, 2132 KiB  
Article
Genomic Insights into Tibetan Sheep Adaptation to Different Altitude Environments
by Wentao Zhang, Chao Yuan, Xuejiao An, Tingting Guo, Caihong Wei, Zengkui Lu and Jianbin Liu
Int. J. Mol. Sci. 2024, 25(22), 12394; https://doi.org/10.3390/ijms252212394 - 19 Nov 2024
Cited by 1 | Viewed by 1595
Abstract
In recent years, research has gradually uncovered the mechanisms of animal adaptation to hypoxic conditions in different altitude environments, particularly at the genomic level. However, past genomic studies on high-altitude adaptation have often not delved deeply into the differences between varying altitude levels. [...] Read more.
In recent years, research has gradually uncovered the mechanisms of animal adaptation to hypoxic conditions in different altitude environments, particularly at the genomic level. However, past genomic studies on high-altitude adaptation have often not delved deeply into the differences between varying altitude levels. This study conducted whole-genome sequencing on 60 Tibetan sheep (Medium Altitude Group (MA): 20 Tao sheep (TS) at 2887 m, High Altitude Group (HA): 20 OuLa sheep (OL) at 3501 m, and Ultra-High Altitude Group (UA): 20 AWang sheep (AW) at 4643 m) from different regions of the Tibetan Plateau in China to assess their responses under varying conditions. Population genetic structure analysis revealed that the three groups are genetically independent, but the TS and OL groups have experienced gene flow with other northern Chinese sheep due to geographical factors. Selection signal analysis identified FGF10, MMP14, SLC25A51, NDUFB8, ALAS1, PRMT1, PRMT5, and HIF1AN as genes associated with ultra-high-altitude hypoxia adaptation, while HMOX2, SEMA4G, SLC16A2, SLC22A17, and BCL2L2 were linked to high-altitude hypoxia adaptation. Functional analysis showed that ultra-high-altitude adaptation genes tend to influence physiological mechanisms directly affecting oxygen uptake, such as lung development, angiogenesis, and red blood cell formation. In contrast, high-altitude adaptation genes are more inclined to regulate mitochondrial DNA replication, iron homeostasis, and calcium signaling pathways to maintain cellular function. Additionally, the functions of shared genes further support the adaptive capacity of Tibetan sheep across a broad geographic range, indicating that these genes offer significant selective advantages in coping with oxygen scarcity. In summary, this study not only reveals the genetic basis of Tibetan sheep adaptation to different altitudinal conditions but also highlights the differences in gene regulation between ultra-high- and high-altitude adaptations. These findings offer new insights into the adaptive evolution of animals in extreme environments and provide a reference for exploring adaptation mechanisms in other species under hypoxic conditions. Full article
(This article belongs to the Special Issue Molecular Genetics and Genomics of Ruminants)
Show Figures

Figure 1

16 pages, 10256 KiB  
Article
PRMT5/WDR77 Enhances the Proliferation of Squamous Cell Carcinoma via the ΔNp63α-p21 Axis
by Heng Liang, Matthew L. Fisher, Caizhi Wu, Carlos Ballon, Xueqin Sun and Alea A. Mills
Cancers 2024, 16(22), 3789; https://doi.org/10.3390/cancers16223789 - 11 Nov 2024
Cited by 2 | Viewed by 1825
Abstract
Protein arginine methyltransferase 5 (PRMT5) is a critical oncogenic factor in various cancers, and its inhibition has shown promise in suppressing tumor growth. However, the role of PRMT5 in squamous cell carcinoma (SCC) remains largely unexplored. In this study, we analyzed SCC patient [...] Read more.
Protein arginine methyltransferase 5 (PRMT5) is a critical oncogenic factor in various cancers, and its inhibition has shown promise in suppressing tumor growth. However, the role of PRMT5 in squamous cell carcinoma (SCC) remains largely unexplored. In this study, we analyzed SCC patient data from The Cancer Genome Atlas (TCGA) and the Cancer Dependency Map (DepMap) to investigate the relationship between PRMT5 and SCC proliferation. We employed competition-based cell proliferation assays, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assays, flow cytometry, and in vivo mouse modeling to examine the regulatory roles of PRMT5 and its binding partner WDR77 (WD repeat domain 77). We identified downstream targets, including the p63 isoform ΔNp63α and the cyclin-dependent kinase inhibitor p21, through single-cell RNA-seq, RT-qPCR, and Western blot analyses. Our findings demonstrate that upregulation of PRMT5 and WDR77 correlates with the poor survival of head and neck squamous cell carcinoma (HNSCC) patients. PRMT5/WDR77 regulates the HNSCC-specific transcriptome and facilitates SCC proliferation by promoting cell cycle progression. The PRMT5 and WDR77 stabilize the ΔNp63α Protein, which in turn, inhibits p21. Moreover, depletion of PRMT5 and WDR77 repress SCC in vivo. This study reveals for the first time that PRMT5 and WDR77 synergize to promote SCC proliferation via the ΔNp63α-p21 axis, highlighting a novel therapeutic target for SCC. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

12 pages, 3309 KiB  
Article
Assessing the Role of AtGRP7 Arginine 141, a Target of Dimethylation by PRMT5, in Flowering Time Control and Stress Response
by Alexander Steffen, Katarzyna Dombert, María José Iglesias, Christine Nolte, María José de Leone, Marcelo J. Yanovsky, Julieta L. Mateos and Dorothee Staiger
Plants 2024, 13(19), 2771; https://doi.org/10.3390/plants13192771 - 3 Oct 2024
Viewed by 1242
Abstract
PROTEIN ARGININE METHYLTRANSFERASES (PRMTs) catalyze arginine (R) methylation that is critical for transcriptional and post-transcriptional gene regulation. In Arabidopsis, PRMT5 that catalyzes symmetric R dimethylation is best characterized. PRMT5 mutants are late-flowering and show altered responses to environmental stress. Among PRMT5 targets are [...] Read more.
PROTEIN ARGININE METHYLTRANSFERASES (PRMTs) catalyze arginine (R) methylation that is critical for transcriptional and post-transcriptional gene regulation. In Arabidopsis, PRMT5 that catalyzes symmetric R dimethylation is best characterized. PRMT5 mutants are late-flowering and show altered responses to environmental stress. Among PRMT5 targets are Arabidopsis thaliana GLYCINE RICH RNA BINDING PROTEIN 7 (AtGRP7) and AtGRP8 that promote the transition to flowering. AtGRP7 R141 has been shown to be modified by PRMT5. Here, we tested whether this symmetric dimethylation of R141 is important for AtGRP7’s physiological role in flowering time control. We constructed AtGRP7 mutant variants with non-methylable R141 (R141A, R141K). Genomic clones containing these variants complemented the late-flowering phenotype of the grp7-1 mutant to the same extent as wild-type AtGRP7. Furthermore, overexpression of AtGRP7 R141A or R141K promoted flowering similar to overexpression of the wild-type protein. Thus, flowering time does not depend on R141 and its modification. However, germination experiments showed that R141 contributes to the activity of AtGRP7 in response to abiotic stress reactions mediated by abscisic acid during early development. Immunoprecipitation of AtGRP7-GFP in the prmt5 background revealed that antibodies against dimethylated arginine still recognized AtGRP7, suggesting that additional methyltransferases may be responsible for modification of AtGRP7. Full article
Show Figures

Figure 1

Back to TopTop