Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.421
Filtrer
1.
Pol J Pathol ; 75(2): 115-125, 2024.
Article de Anglais | MEDLINE | ID: mdl-39166520

RÉSUMÉ

Precancerous cervical lesions are metaplastic alterations of epithelial cells of the cervix, eventually developing into cervical cancer. Despite primary and secondary prevention, the burden of cervical cancer remains high globally. Protein arginine methyltransferases (PRMT) represent post-translational modifications that interact with multiple signalling pathways, playing a role in epithelial-mesenchymal transition. In complex with desmoglein-2 (DSG2), a cell adhesion protein, both participate in the progression of dysplastic changes with potential malignant development. The presented study was performed on archival paraffin-embedded blocks from adult women. The studied samples were categorised into low-grade and high-grade intraepithelial lesions. Immunohistochemical analysis was used to observe subcellular localisation, immunoreaction intensity, and percentage of PRMT5- and DSG2-expressing cells, followed by statistical analysis. Preliminary results identified statistically significant differences between the expression and subcellular localisation of proteins in question in low-grade and high-grade squamous intraepithelial lesions. The primary goal of the presented study is to perceive the involvement of PRMT5 and DSG2 in the initiation and progression of cervical lesions. Our observations indicate the potential of the assessed proteins as prognostic markers. However, further studies of PRMT5 and DSG2 are required to provide greater insight into cervical carcinogenesis.


Sujet(s)
Marqueurs biologiques tumoraux , Desmogléine-2 , États précancéreux , Protein-arginine N-methyltransferases , Tumeurs du col de l'utérus , Humains , Femelle , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/métabolisme , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/analyse , Desmogléine-2/métabolisme , Desmogléine-2/analyse , Marqueurs biologiques tumoraux/analyse , Marqueurs biologiques tumoraux/métabolisme , États précancéreux/anatomopathologie , États précancéreux/métabolisme , Adulte , Pronostic , Adulte d'âge moyen , Immunohistochimie , Dysplasie du col utérin/anatomopathologie , Dysplasie du col utérin/métabolisme
2.
Int J Mol Sci ; 25(16)2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39201539

RÉSUMÉ

Breast cancer is the most common cancer diagnosed in women worldwide. Early-stage breast cancer is curable in ~70-80% of patients, while advanced metastatic breast cancer is considered incurable with current therapies. Breast cancer is a highly heterogeneous disease categorized into three main subtypes based on key markers orientating specific treatment strategies for each subtype. The complexity of breast carcinogenesis is often associated with epigenetic modification regulating different signaling pathways, involved in breast tumor initiation and progression, particularly by the methylation of arginine residues. Protein arginine methyltransferases (PRMT1-9) have emerged, through their ability to methylate histones and non-histone substrates, as essential regulators of cancers. Here, we present an updated overview of the mechanisms by which PRMT1 and PRMT5, two major members of the PRMT family, control important signaling pathways impacting breast tumorigenesis, highlighting them as putative therapeutic targets.


Sujet(s)
Tumeurs du sein , Protein-arginine N-methyltransferases , Protéines de répression , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Femelle , Protéines de répression/métabolisme , Protéines de répression/génétique , Transduction du signal , Méthylation , Épigenèse génétique , Animaux , Régulation de l'expression des gènes tumoraux
3.
Int J Biol Macromol ; 277(Pt 3): 134411, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39097054

RÉSUMÉ

Stress granules (SGs) are membrane-less organelles (MLOs) or cytosolic compartments formed upon exposure to environmental cell stress-inducing stimuli. SGs are based on ribonucleoprotein complexes from a set of cytoplasmic proteins and mRNAs, blocked in translation due to stress cell-induced polysome disassembly. Post-translational modifications (PTMs) such as methylation, are involved in SG assembly, with the methylation writer PRMT1 and its reader TDRD3 colocalizing to SGs. However, the role of this writer-reader system in SG assembly remains unclear. Here, we found that PRMT1 methylates SG constituent RNA-binding proteins (RBPs) on their RGG motifs. Besides, we report that TDRD3, as a reader of asymmetric dimethylarginines, enhances RNA binding to recruit additional RNAs and RBPs, lowering the percolation threshold and promoting SG assembly. Our study enriches our understanding of the molecular mechanism of SG formation by elucidating the functions of PRMT1 and TDRD3. We anticipate that our study will provide a new perspective for comprehensively understanding the functions of PTMs in liquid-liquid phase separation driven condensate assembly.


Sujet(s)
Protein-arginine N-methyltransferases , Protéines de liaison à l'ARN , Granules de stress , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Humains , Méthylation , Granules de stress/métabolisme , Protéines de liaison à l'ARN/métabolisme , Cartes d'interactions protéiques , Protéines de répression/métabolisme , Protéines de répression/génétique , Liaison aux protéines , Maturation post-traductionnelle des protéines
4.
Cancer Lett ; 601: 217148, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39098759

RÉSUMÉ

Studying the mechanisms underlying clear cell renal cell carcinoma (ccRCC), the most common subtype of kidney cancer, may address an unmet need in ccRCC-targeted drug research. Growing evidences indicate that protein phosphatase 4 (PP4) plays an important role in cancer biology. Here, we characterized the upregulation of PP4 core component SMEK1 in ccRCC using tissue microarrays and revealed that its high expression is closely associated with reduced patient survival. We then conducted cell function experiments and animal experiments to prove the tumor-promoting effect of SMEK1. Next, RNA-seq was performed to explore its underlying mechanism, and the results revealed that SMEK1-regulated genes were extensively involved in cell motility, and the canonical tyrosine kinase receptor EGFR was one of its targets. Moreover, we verified the regulatory effect of SMEK1 on EGFR and its downstream MAPK and AKT pathway through molecular experiments, in which erlotinib, a tyrosine kinase inhibitor, can partially block this regulation, demonstrating that SMEK1 mediates its effects dependent on the tyrosine kinase activity of EGFR. Mechanistically, SMEK1 bond to PRMT5 and facilitated PRMT5-mediated histone methylation to promote the transcription of EGFR. Furthermore, we studied the upstream regulators of SMEK1 and demonstrated that the transcription factor E2F1 could directly bind to the SMEK1 promoter by chromatin immunoprecipitation. Functionally, E2F1 could also induce ccRCC progression by manipulating the expression of SMEK1. Collectively, our findings demonstrate the overexpression of SMEK1 in ccRCC, and reveal a novel E2F1/SMEK1/PRMT5/EGFR-tyrosine-kinase-dependent pathway for ccRCC progression.


Sujet(s)
Néphrocarcinome , Évolution de la maladie , Récepteurs ErbB , Régulation de l'expression des gènes tumoraux , Tumeurs du rein , Humains , Néphrocarcinome/anatomopathologie , Néphrocarcinome/génétique , Néphrocarcinome/métabolisme , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Tumeurs du rein/anatomopathologie , Tumeurs du rein/génétique , Tumeurs du rein/métabolisme , Animaux , Lignée cellulaire tumorale , Souris , Transduction du signal , Mouvement cellulaire , Mâle , Protein-arginine N-methyltransferases/génétique , Protein-arginine N-methyltransferases/métabolisme , Femelle , Facteur de transcription E2F1/métabolisme , Facteur de transcription E2F1/génétique
5.
Biochemistry ; 63(17): 2141-2152, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39146246

RÉSUMÉ

Dipeptide repeat proteins (DPRs) are aberrant protein species found in C9orf72-linked amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two neurodegenerative diseases characterized by the cytoplasmic mislocalization and aggregation of RNA-binding proteins (RBPs). In particular, arginine (R)-rich DPRs (poly-GR and poly-PR) have been suggested to promiscuously interact with multiple cellular proteins and thereby exert high cytotoxicity. Components of the protein arginine methylation machinery have been identified as modulators of DPR toxicity and/or potential cellular interactors of R-rich DPRs; however, the molecular details and consequences of such an interaction are currently not well understood. Here, we demonstrate that several members of the family of protein arginine methyltransferases (PRMTs) can directly interact with R-rich DPRs in vitro and in the cytosol. In vitro, R-rich DPRs reduce solubility and promote phase separation of PRMT1, the main enzyme responsible for asymmetric arginine-dimethylation (ADMA) in mammalian cells, in a concentration- and length-dependent manner. Moreover, we demonstrate that poly-GR interferes more efficiently than poly-PR with PRMT1-mediated arginine methylation of RBPs such as hnRNPA3. We additionally show by two alternative approaches that poly-GR itself is a substrate for PRMT1-mediated arginine dimethylation. We propose that poly-GR may act as a direct competitor for arginine methylation of cellular PRMT1 targets, such as disease-linked RBPs.


Sujet(s)
Arginine , Protein-arginine N-methyltransferases , Protéines de liaison à l'ARN , Protéines de répression , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Humains , Arginine/métabolisme , Méthylation , Protéines de répression/métabolisme , Protéines de répression/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Sclérose latérale amyotrophique/métabolisme , Sclérose latérale amyotrophique/génétique , Démence frontotemporale/métabolisme , Démence frontotemporale/génétique , Protéine C9orf72/métabolisme , Protéine C9orf72/génétique , Cellules HEK293
6.
Theranostics ; 14(10): 4090-4106, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994016

RÉSUMÉ

Purpose: Due to intrinsic defensive response, ferroptosis-activating targeted therapy fails to achieve satisfactory clinical benefits. Though p62-Keap1-Nrf2 axis is activated to form a negative feedback loop during ferroptosis induction, how p62 is activated remains largely unknown. Methods: MTS assay was applied to measure cell growth. Lipid ROS was detected with C11-BODIPY reagent by flow cytometer. Quantitative real-time PCR (qPCR) and western blotting were performed to determine mRNA and protein level. Immunofluorescence (IF) was performed to examine the distribution of proteins. Fluorescence recovery after photobleaching (FRAP) was adopted to evaluate p62 phase separation. Immunoprecipitation (IP), co-IP and Proximal ligation assay (PLA) were performed to detected protein posttranslational modifications and protein-protein interactions. Tumor xenograft model was employed to inspect in vivo growth of pancreatic cancer cells. Results: Upon ferroptosis induction, Nuclear Factor E2 Related Factor 2 (Nrf2) protein and its downstream genes such as HMOX1 and NQO1 were upregulated. Knockdown of p62 significantly reversed Nrf2 upregulation and Keap1 decrease after ferroptosis induction. Knockdown of either p62 or Nrf2 remarkably sensitized ferroptosis induction. Due to augmented p62 phase separation, formation of p62 bodies were increased to recruit Keap1 after ferroptosis induction. Protein arginine methyltransferase 6 (PRMT6) mediated asymmetric dimethylarginine (ADMA) of p62 to increase its oligomerization, promoting p62 phase separation and p62 body formation. Knockdown of p62 or PRMT6 notably sensitized pancreatic cancer cells to ferroptosis both in vitro and in vivo through suppressing Nrf2 signaling. Conclusion: During ferroptosis induction, PRMT6 mediated p62 ADMA to promote its phase separation, sequestering Keap1 to activate Nrf2 signaling and inhibit ferroptosis. Therefore, targeting PRMT6-mediated p62 ADMA could be a new option to sensitize ferroptosis for cancer treatment.


Sujet(s)
Arginine , Ferroptose , Protéine-1 de type kelch associée à ECH , Facteur-2 apparenté à NF-E2 , Protein-arginine N-methyltransferases , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Humains , Animaux , Arginine/métabolisme , Arginine/analogues et dérivés , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Souris , Lignée cellulaire tumorale , Protéine-1 de type kelch associée à ECH/métabolisme , Protéine-1 de type kelch associée à ECH/génétique , Rétrocontrôle physiologique , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Séquestosome-1/métabolisme , Séquestosome-1/génétique , Souris nude , Transduction du signal , , Protéines de liaison à l'ARN
7.
Protein Sci ; 33(8): e5118, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39022984

RÉSUMÉ

Proper protein arginine methylation by protein arginine methyltransferase 1 (PRMT1) is critical for maintaining cellular health, while dysregulation is often associated with disease. How the activity of PRMT1 is regulated is therefore paramount, but is not clearly understood. Several studies have observed higher order oligomeric species of PRMT1, but it is unclear if these exist at physiological concentrations and there is confusion in the literature about how oligomerization affects activity. We therefore sought to determine which oligomeric species of PRMT1 are physiologically relevant, and quantitatively correlate activity with specific oligomer forms. Through quantitative western blotting, we determined that concentrations of PRMT1 available in a variety of human cell lines are in the sub-micromolar to low micromolar range. Isothermal spectral shift binding data were modeled to a monomer/dimer/tetramer equilibrium with an EC50 for tetramer dissociation of ~20 nM. A combination of sedimentation velocity and Native polyacrylamide gel electrophoresis experiments directly confirmed that the major oligomeric species of PRMT1 at physiological concentrations would be dimers and tetramers. Surprisingly, the methyltransferase activity of a dimeric PRMT1 variant is similar to wild type, tetrameric PRMT1 with some purified substrates, but dimer and tetramer forms of PRMT1 show differences in catalytic efficiencies and substrate specificity for other substrates. Our results define an oligomerization paradigm for PRMT1, show that the biophysical characteristics of PRMT1 are poised to support a monomer/dimer/tetramer equilibrium in vivo, and suggest that the oligomeric state of PRMT1 could be used to regulate substrate specificity.


Sujet(s)
Multimérisation de protéines , Protein-arginine N-methyltransferases , Protein-arginine N-methyltransferases/composition chimique , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Humains , Spécificité du substrat , Protéines de répression/composition chimique , Protéines de répression/métabolisme , Protéines de répression/génétique , Méthylation
8.
Cell Death Dis ; 15(7): 524, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39043634

RÉSUMÉ

Invasion and migration are the key hallmarks of cancer, and aggressive growth is a major factor contributing to treatment failure and poor prognosis in glioblastoma. Protein arginine methyltransferase 6 (PRMT6), as an epigenetic regulator, has been confirmed to promote the malignant proliferation of glioblastoma cells in previous studies. However, the effects of PRMT6 on glioblastoma cell invasion and migration and its underlying mechanisms remain elusive. Here, we report that PRMT6 functions as a driver element for tumor cell invasion and migration in glioblastoma. Bioinformatics analysis and glioma sample detection results demonstrated that PRMT6 is highly expressed in mesenchymal subtype or invasive gliomas, and is significantly negatively correlated with their prognosis. Inhibition of PRMT6 (using PRMT6 shRNA or inhibitor EPZ020411) reduces glioblastoma cell invasion and migration in vitro, whereas overexpression of PRMT6 produces opposite effects. Then, we identified that PRMT6 maintains the protein stability of EZH2 by inhibiting the degradation of EZH2 protein, thereby mediating the invasion and migration of glioblastoma cells. Further mechanistic investigations found that PRMT6 inhibits the transcription of TRAF6 by activating the histone methylation mark (H3R2me2a), and reducing the interaction between TRAF6 and EZH2 to enhance the protein stability of EZH2 in glioblastoma cells. Xenograft tumor assay and HE staining results showed that the expression of PRMT6 could promote the invasion of glioblastoma cells in vivo, the immunohistochemical staining results of mouse brain tissue tumor sections also confirmed the regulatory relationship between PRMT6, TRAF6, and EZH2. Our findings illustrate that PRMT6 suppresses TRAF6 transcription via H3R2me2a to enhance the protein stability of EZH2 to facilitate glioblastoma cell invasion and migration. Blocking the PRMT6-TRAF6-EZH2 axis is a promising strategy for inhibiting glioblastoma cell invasion and migration.


Sujet(s)
Mouvement cellulaire , Protéine-2 homologue de l'activateur de Zeste , Glioblastome , Invasion tumorale , Stabilité protéique , Protein-arginine N-methyltransferases , Ubiquitination , Animaux , Femelle , Humains , Mâle , Souris , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Lignée cellulaire tumorale , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Régulation de l'expression des gènes tumoraux , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Glioblastome/génétique , Souris de lignée BALB C , Souris nude , Protéines nucléaires , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Protéolyse , Facteur-6 associé aux récepteurs de TNF/métabolisme , Facteur-6 associé aux récepteurs de TNF/génétique
9.
J Med Chem ; 67(16): 13604-13638, 2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39080842

RÉSUMÉ

PRMT5, a type 2 arginine methyltransferase, has a critical role in regulating cell growth and survival in cancer. With the aim of developing MTA-cooperative PRMT5 inhibitors suitable for MTAP-deficient cancers, herein we report our efforts to develop novel "MTA-cooperative" compounds identified through a high-throughput biochemical screening approach. Optimization of hits was achieved through structure-based design with a focus on improvement of oral drug-like properties. Bioisosteric replacement of the original thiazole guanidine headgroup, spirocyclization of the isoindolinone amide scaffold to both configurationally and conformationally lock the bioactive form, and fine-tuning of the potency, MTA cooperativity, and DMPK properties through specific substitutions of the azaindole headgroup were conducted. We have identified an orally available in vivo lead compound, 28 ("AZ-PRMT5i-1"), which shows sub-10 nM PRMT5 cell potency, >50-fold MTA cooperativity, suitable DMPK properties for oral dosing, and significant PRMT5-driven in vivo efficacy in several MTAP-deficient preclinical cancer models.


Sujet(s)
Antienzymes , Protein-arginine N-methyltransferases , Protein-arginine N-methyltransferases/antagonistes et inhibiteurs , Protein-arginine N-methyltransferases/métabolisme , Humains , Animaux , Antienzymes/pharmacologie , Antienzymes/composition chimique , Antienzymes/synthèse chimique , Relation structure-activité , Souris , Découverte de médicament , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique
10.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(3): 349-358, 2024 Mar 28.
Article de Anglais, Chinois | MEDLINE | ID: mdl-38970508

RÉSUMÉ

OBJECTIVES: Obesity related glomerulopathy (ORG) is induced by obesity, but the pathogenesis remains unclear. This study aims to investigate the expression of early growth response protein 3 (EGR3) in the renal cortex tissues of ORG patients and high-fat diet-induced obese mice, and to further explore the molecular mechanism of EGR3 in inhibiting palmitic acid (PA) induced human podocyte inflammatory damage. METHODS: Renal cortex tissues were collected from ORG patients (n=6) who have been excluded from kidney damage caused by other diseases and confirmed by histopathology, and from obese mice induced by high-fat diet (n=10). Human and mouse podocytes were intervened with 150 µmol/L PA for 48 hours. EGR3 was overexpressed or silenced in human podocytes. Enzyme linked immunosorbent assay (ELISA) was used to detcet the levels of interleukin-6 (IL-6) and interleukin-1ß (IL-1ß). Real-time RT-PCR was used to detect the mRNA expressions of EGR3, podocytes molecular markers nephrosis 1 (NPHS1), nephrosis 2 (NPHS2), podocalyxin (PODXL), and podoplanin (PDPN). RNA-seq was performed to detect differentially expressed genes (DEGs) after human podocytes overexpressing EGR3 and treated with 150 µmol/L PA compared with the control group. Co-immunoprecipitation (Co-IP) combined with liquid chromatography tandem mass spectrometry (LC-MS) was used to detect potential interacting proteins of EGR3 and the intersected with the RNA-seq results. Co-IP confirmed the interaction between EGR3 and protein arginine methyltransferases 1 (PRMT1), after silencing EGR3 and PRMT1 inhibitor intervention, the secretion of IL-6 and IL-1ß in PA-induced podocytes was detected. Western blotting was used to detect the expression of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) after overexpression or silencing of EGR3. RESULTS: EGR3 was significantly upregulated in renal cortex tissues of ORG patients and high-fat diet-induced obese mice (both P<0.01). In addition, after treating with 150 µmol/L PA for 48 hours, the expression of EGR3 in human and mouse podocytes was significantly upregulated (both P<0.05). Overexpression or silencing of EGR3 in human podocytes inhibited or promoted the secretion of IL-6 and IL-1ß in the cell culture supernatant after PA intervention, respectively, and upregulated or downregulated the expression of NPHS1, PODXL, NPHS2,and PDPN (all P<0.05). RNA-seq showed a total of 988 DEGs, and Co-IP+LC-MS identified a total of 238 proteins that may interact with EGR3. Co-IP confirmed that PRMT1 was an interacting protein with EGR3. Furthermore, PRMT1 inhibitors could partially reduce PA-induced IL-6 and IL-1ß secretion after EGR3 silencing in human podocytes (both P<0.05). Overexpression or silencing of EGR3 negatively regulated the expression of PRMT1 and p-STAT3. CONCLUSIONS: EGR3 may reduce ORG podocyte inflammatory damage by inhibiting the PRMT1/p-STAT3 pathway.


Sujet(s)
Facteur de transcription EGR-3 , Obésité , Podocytes , Protein-arginine N-methyltransferases , Protéines de répression , Facteur de transcription STAT-3 , Animaux , Humains , Mâle , Souris , Alimentation riche en graisse/effets indésirables , Facteur de transcription EGR-3/métabolisme , Facteur de transcription EGR-3/génétique , Inflammation/métabolisme , Interleukine-1 bêta/métabolisme , Interleukine-6/métabolisme , Interleukine-6/génétique , Cortex rénal/métabolisme , Cortex rénal/anatomopathologie , Maladies du rein/métabolisme , Maladies du rein/étiologie , Maladies du rein/anatomopathologie , Souris de lignée C57BL , Souris obèse , Obésité/complications , Obésité/métabolisme , Acide palmitique/pharmacologie , Podocytes/métabolisme , Podocytes/anatomopathologie , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Protéines de répression/métabolisme , Protéines de répression/génétique , Transduction du signal , Facteur de transcription STAT-3/métabolisme
11.
Diabetes Metab J ; 48(4): 487-502, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39043443

RÉSUMÉ

Cardiovascular diseases (CVDs) and metabolic disorders stand as formidable challenges that significantly impact the clinical outcomes and living quality for afflicted individuals. An intricate comprehension of the underlying mechanisms is paramount for the development of efficacious therapeutic strategies. Protein arginine methyltransferases (PRMTs), a class of enzymes responsible for the precise regulation of protein methylation, have ascended to pivotal roles and emerged as crucial regulators within the intrinsic pathophysiology of these diseases. Herein, we review recent advancements in research elucidating on the multifaceted involvements of PRMTs in cardiovascular system and metabolic diseases, contributing significantly to deepen our understanding of the pathogenesis and progression of these maladies. In addition, this review provides a comprehensive analysis to unveil the distinctive roles of PRMTs across diverse cell types implicated in cardiovascular and metabolic disorders, which holds great potential to reveal novel therapeutic interventions targeting PRMTs, thus presenting promising perspectives to effectively address the substantial global burden imposed by CVDs and metabolic disorders.


Sujet(s)
Maladies cardiovasculaires , Maladies métaboliques , Protein-arginine N-methyltransferases , Humains , Maladies cardiovasculaires/métabolisme , Maladies métaboliques/métabolisme , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/antagonistes et inhibiteurs , Animaux , Méthylation
12.
Sci Rep ; 14(1): 17316, 2024 07 27.
Article de Anglais | MEDLINE | ID: mdl-39068290

RÉSUMÉ

PRMT5 is a widely expressed arginine methyltransferase that regulates processes involved in tumor cell proliferation and survival. In the study described here, we investigated whether PRMT5 provides a target for tumor radiosensitization. Knockdown of PRMT5 using siRNA enhanced the radiosensitivity of a panel of cell lines corresponding to tumor types typically treated with radiotherapy. To extend these studies to an experimental therapeutic setting, the PRMT5 inhibitor LLY-283 was used. Exposure of the tumor cell lines to LLY-283 decreased PRMT5 activity and enhanced their radiosensitivity. This increase in radiosensitivity was accompanied by an inhibition of DNA double-strand break repair as determined by γH2AX foci and neutral comet analyses. For a normal fibroblast cell line, although LLY-283 reduced PRMT5 activity, it had no effect on their radiosensitivity. Transcriptome analysis of U251 cells showed that LLY-283 treatment reduced the expression of genes and altered the mRNA splicing pattern of genes involved in the DNA damage response. Subcutaneous xenografts were then used to evaluate the in vivo response to LLY-283 and radiation. Treatment of mice with LLY-283 decreased tumor PRMT5 activity and significantly enhanced the radiation-induced growth delay. These results suggest that PRMT5 is a tumor selective target for radiosensitization.


Sujet(s)
Protein-arginine N-methyltransferases , Radiotolérance , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Protein-arginine N-methyltransferases/antagonistes et inhibiteurs , Animaux , Humains , Radiotolérance/effets des médicaments et des substances chimiques , Radiotolérance/génétique , Lignée cellulaire tumorale , Souris , Réparation de l'ADN , Prolifération cellulaire/effets des radiations , Tests d'activité antitumorale sur modèle de xénogreffe , Cassures double-brin de l'ADN/effets des radiations , Souris nude
13.
Poult Sci ; 103(9): 103989, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38981362

RÉSUMÉ

Protein arginine methyltransferases (PRMTs) in mammals play a role in various signaling pathways, such as virus infection, inflammasome responses, and cancer growth. While some PRMTs have been found to regulate interferon production in mammals, the mechanism in chickens remains to be fully understood. This study focused on investigating the function of chicken PRMTs. Our findings indicate that chicken PRMTs act as inhibitors of interferon production in response to dsRNA or MDA5 stimulation. Each PRMT is involved in different stages of interferon induction through the MDA5-MAVS-TBK1 pathway. Furthermore, we observed the colocalization of multiple PRMTs with the viral protein VP3 of infectious bursal disease virus (IBDV). Among the chicken PRMTs studied, PRMT3 was found to be widely expressed in various organs and its expression was upregulated during IBDV infection. Notably, PRMT3 supported IBDV replication, as demonstrated by ectopic expression and inhibition studies using SGC-707. Silencing of PRMT3 led to enhanced interferon production and inhibition of IBDV replication. This study provides novel insights into the role of chicken PRMTs, particularly PRMT3, in promoting IBDV replication by suppressing interferon signaling.


Sujet(s)
Protéines aviaires , Poulets , Virus de la bursite infectieuse , Maladies de la volaille , Protein-arginine N-methyltransferases , Réplication virale , Animaux , Virus de la bursite infectieuse/physiologie , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Protéines aviaires/génétique , Protéines aviaires/métabolisme , Maladies de la volaille/virologie , Infections à Birnaviridae/médecine vétérinaire , Infections à Birnaviridae/virologie
14.
Int J Mol Sci ; 25(14)2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39063139

RÉSUMÉ

Non-alcoholic fatty liver disease (NAFLD) has emerged as the leading cause of chronic liver disease worldwide. Caspase 8 and FADD-like apoptosis regulator (CFLAR) has been identified as a potent factor in mitigating non-alcoholic steatohepatitis (NASH) by inhibiting the N-terminal dimerization of apoptosis signal-regulating kinase 1 (ASK1). While arginine methyltransferase 1 (PRMT1) was previously reported to be associated with increased hepatic glucose production, its involvement in hepatic lipid metabolism remains largely unexplored. The interaction between PRMT1 and CFLAR and the methylation of CFLAR were verified by Co-IP and immunoblotting assays. Recombinant adenoviruses were generated for overexpression or knockdown of PRMT1 in hepatocytes. The role of PRMT1 in NAFLD was investigated in normal and high-fat diet-induced obese mice. In this study, we found a significant upregulation of PRMT1 and downregulation of CFLAR after 48h of fasting, while the latter significantly rebounded after 12h of refeeding. The expression of PRMT1 increased in the livers of mice fed a methionine choline-deficient (MCD) diet and in hepatocytes challenged with oleic acid (OA)/palmitic acid (PA). Overexpression of PRMT1 not only inhibited the expression of genes involved in fatty acid oxidation (FAO) and promoted the expression of genes involved in fatty acid synthesis (FAS), resulting in increased triglyceride accumulation in primary hepatocytes, but also enhanced the gluconeogenesis of primary hepatocytes. Conversely, knockdown of hepatic PRMT1 significantly alleviated MCD diet-induced hepatic lipid metabolism abnormalities and liver injury in vivo, possibly through the upregulation of CFLAR protein levels. Knockdown of PRMT1 suppressed the expression of genes related to FAS and enhanced the expression of genes involved in FAO, causing decreased triglyceride accumulation in OA/PA-treated primary hepatocytes in vitro. Although short-term overexpression of PRMT1 had no significant effect on hepatic triglyceride levels under physiological conditions, it resulted in increased serum triglyceride and fasting blood glucose levels in normal C57BL/6J mice. More importantly, PRMT1 was observed to interact with and methylate CFLAR, ultimately leading to its ubiquitination-mediated protein degradation. This process subsequently triggered the activation of c-Jun N-terminal kinase 1 (JNK1) and lipid deposition in primary hepatocytes. Together, these results suggested that PRMT1-mediated methylation of CFLAR plays a critical role in hepatic lipid metabolism. Targeting PRMT1 for drug design may represent a promising strategy for the treatment of NAFLD.


Sujet(s)
Hépatocytes , Métabolisme lipidique , Foie , Stéatose hépatique non alcoolique , Protein-arginine N-methyltransferases , Animaux , Humains , Mâle , Souris , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/génétique , Alimentation riche en graisse/effets indésirables , Hépatocytes/métabolisme , Foie/métabolisme , Méthylation , Souris de lignée C57BL , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/génétique , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique
15.
J Mol Biol ; 436(19): 168727, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39079611

RÉSUMÉ

Stress granules (SGs) are dynamic biomolecular condensates that form in the cytoplasm in response to cellular stress, encapsulating proteins and RNAs. Methylation is a key factor in the assembly of SGs, with PRMT1, which acts as an arginine methyltransferase, localizing to SGs. However, the precise mechanism of PRMT1 localization within SGs remains unknown. In this study, we identified that Caprin1 plays a primary role in the recruitment of PRMT1 to SGs, particularly through its C-terminal domain. Our findings demonstrate that Caprin1 serves a dual function as both a linker, facilitating the formation of a PRMT1-G3BP1 complex, and as a spacer, preventing the aberrant formation of SGs under non-stress conditions. This study sheds new lights on the regulatory mechanisms governing SG formation and suggests that Caprin1 plays a critical role in cellular responses to stress.


Sujet(s)
Protéines du cycle cellulaire , Helicase , Protéines liant le poly-adp-ribose , Protein-arginine N-methyltransferases , RNA helicases , Protéines à motif de reconnaissance de l'ARN , Protéines de répression , Granules de stress , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Protéines à motif de reconnaissance de l'ARN/métabolisme , Protéines à motif de reconnaissance de l'ARN/génétique , Protéines liant le poly-adp-ribose/métabolisme , Protéines liant le poly-adp-ribose/génétique , RNA helicases/métabolisme , RNA helicases/génétique , Humains , Helicase/métabolisme , Helicase/génétique , Granules de stress/métabolisme , Protéines de répression/métabolisme , Protéines de répression/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Liaison aux protéines , Méthylation , Granulations cytoplasmiques/métabolisme , Cellules HeLa
16.
Cell Rep ; 43(7): 114459, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38985674

RÉSUMÉ

Glycine- and arginine-rich (GAR) motifs, commonly found in RNA-binding and -processing proteins, can be symmetrically (SDMA) or asymmetrically (ADMA) dimethylated at the arginine residue by protein arginine methyltransferases. Arginine-methylated protein motifs are usually read by Tudor domain-containing proteins. Here, using a GFP-Trap, we identify a non-Tudor domain protein, squamous cell carcinoma antigen recognized by T cells 3 (SART3), as a reader for SDMA-marked GAR motifs. Structural analysis and mutagenesis of SART3 show that aromatic residues lining a groove between two adjacent aromatic-rich half-a-tetratricopeptide (HAT) repeat domains are essential for SART3 to recognize and bind to SDMA-marked GAR motif peptides, as well as for the interaction between SART3 and the GAR-motif-containing proteins fibrillarin and coilin. Further, we show that the loss of this reader ability affects RNA splicing. Overall, our findings broaden the range of potential SDMA readers to include HAT domains.


Sujet(s)
Motifs d'acides aminés , Arginine , Glycine , Arginine/métabolisme , Arginine/composition chimique , Humains , Glycine/métabolisme , Glycine/composition chimique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/composition chimique , Protéines de liaison à l'ARN/génétique , Liaison aux protéines , Épissage des ARN , Cellules HEK293 , Méthylation , Protéines chromosomiques nonhistones/métabolisme , Protéines chromosomiques nonhistones/composition chimique , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/composition chimique
17.
Cell Death Dis ; 15(7): 504, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009589

RÉSUMÉ

Abnormal epigenetic modifications are involved in the regulation of Warburg effect in tumor cells. Protein arginine methyltransferases (PRMTs) mediate arginine methylation and have critical functions in cellular responses. PRMTs are deregulated in a variety of cancers, but their precise roles in Warburg effect in cancer is largely unknown. Experiments from the current study showed that PRMT1 was highly expressed under conditions of glucose sufficiency. PRMT1 induced an increase in the PKM2/PKM1 ratio through upregulation of PTBP1, in turn, promoting aerobic glycolysis in non-small cell lung cancer (NSCLC). The PRMT1 level in p53-deficient and p53-mutated NSCLC remained relatively unchanged while the expression was reduced in p53 wild-type NSCLC under conditions of glucose insufficiency. Notably, p53 activation under glucose-deficient conditions could suppress USP7 and further accelerate the polyubiquitin-dependent degradation of PRMT1. Melatonin, a hormone that inhibits glucose intake, markedly suppressed cell proliferation of p53 wild-type NSCLC, while a combination of melatonin and the USP7 inhibitor P5091 enhanced the anticancer activity in p53-deficient NSCLC. Our collective findings support a role of PRMT1 in the regulation of Warburg effect in NSCLC. Moreover, combination treatment with melatonin and the USP7 inhibitor showed good efficacy, providing a rationale for the development of PRMT1-based therapy to improve p53-deficient NSCLC outcomes.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Protéines membranaires , Protein-arginine N-methyltransferases , , Hormones thyroïdiennes , Protéine p53 suppresseur de tumeur , Effet Warburg en oncologie , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Humains , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Effet Warburg en oncologie/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Hormones thyroïdiennes/métabolisme , Lignée cellulaire tumorale , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines de transport/métabolisme , Protéines de transport/génétique , Ubiquitin-specific peptidase 7/métabolisme , Ubiquitin-specific peptidase 7/génétique , Protéines de répression/métabolisme , Protéines de répression/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Animaux , Glycolyse/effets des médicaments et des substances chimiques , Souris nude , Glucose/métabolisme , Souris , Régulation de l'expression des gènes tumoraux , Cellules A549 , Protéine PTB
18.
Genes (Basel) ; 15(6)2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38927636

RÉSUMÉ

Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia/lymphoma. The oncogene product Tax of HTLV-I is thought to play crucial roles in leukemogenesis by promoting proliferation of the virus-infected cells through activation of growth-promoting genes. These genes code for growth factors and their receptors, cytokines, cell adhesion molecules, growth signal transducers, transcription factors and cell cycle regulators. We show here that Tax activates the gene coding for coactivator-associated arginine methyltransferase 1 (CARM1), which epigenetically enhances gene expression through methylation of histones. Tax activated the Carm1 gene and increased protein expression, not only in human T-cell lines but also in normal peripheral blood lymphocytes (PHA-PBLs). Tax increased R17-methylated histone H3 on the target gene IL-2Rα, concomitant with increased expression of CARM1. Short hairpin RNA (shRNA)-mediated knockdown of CARM1 decreased Tax-mediated induction of IL-2Rα and Cyclin D2 gene expression, reduced E2F activation and inhibited cell cycle progression. Tax acted via response elements in intron 1 of the Carm1 gene, through the NF-κB pathway. These results suggest that Tax-mediated activation of the Carm1 gene contributes to leukemogenic target-gene expression and cell cycle progression, identifying the first epigenetic target gene for Tax-mediated trans-activation in cell growth promotion.


Sujet(s)
Produits du gène tax , Virus T-lymphotrope humain de type 1 , Protein-arginine N-methyltransferases , Humains , Protein-arginine N-methyltransferases/génétique , Protein-arginine N-methyltransferases/métabolisme , Produits du gène tax/génétique , Produits du gène tax/métabolisme , Virus T-lymphotrope humain de type 1/génétique , Cycline D2/génétique , Cycline D2/métabolisme , Activation de la transcription , Sous-unité alpha du récepteur à l'interleukine-2/génétique , Sous-unité alpha du récepteur à l'interleukine-2/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/génétique , Histone/métabolisme , Histone/génétique , Épigenèse génétique , Cellules Jurkat
19.
Int J Mol Sci ; 25(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38892217

RÉSUMÉ

Microglia-mediated inflammatory response is one key cause of many central nervous system diseases, like Alzheimer's disease. We hypothesized that a novel C15orf39 (MAPK1 substrate) plays a critical role in the microglial inflammatory response. To confirm this hypothesis, we used lipopolysaccharide (LPS)-and interferon-gamma (IFN-γ)-induced human microglia HMC3 cells as a representative indicator of the microglial in vitro inflammatory response. We found that C15orf39 was down-regulated when interleukin-6 (IL-6) and tumor necrosis factor-α (TNFα) expression increased in LPS/IFN-γ-stimulated HMC3 cells. Once C15orf39 was overexpressed, IL-6 and TNFα expression were reduced in LPS/IFN-γ-stimulated HMC3 cells. In contrast, C15orf39 knockdown promoted IL-6 and TNFα expression in LPS/IFN-γ-stimulated HMC3 cells. These results suggest that C15orf39 is a suppressive factor in the microglial inflammatory response. Mechanistically, C15orf39 interacts with the cytoplasmic protein arginine methyltransferase 2 (PRMT2). Thus, we termed C15orf39 a PRMT2 interaction protein (PRMT2 IP). Furthermore, the interaction of C15orf39 and PRMT2 suppressed the activation of NF-κB signaling via the PRMT2-IκBα signaling axis, which then led to a reduction in transcription of the inflammatory factors IL6 and TNF-α. Under inflammatory conditions, NF-κBp65 was found to be activated and to suppress C15orf39 promoter activation, after which it canceled the suppressive effect of the C15orf39-PRMT2-IκBα signaling axis on IL-6 and TNFα transcriptional expression. In conclusion, our findings demonstrate that in a steady condition, the interaction of C15orf39 and PRMT2 stabilizes IκBα to inhibit IL-6 and TNFα expression by suppressing NF-κB signaling, which reversely suppresses C15orf39 transcription to enhance IL-6 and TNFα expression in the microglial inflammatory condition. Our study provides a clue as to the role of C15orf39 in microglia-mediated inflammation, suggesting the potential therapeutic efficacy of C15orf39 in some central nervous system diseases.


Sujet(s)
Inflammation , Interleukine-6 , Lipopolysaccharides , Microglie , Protein-arginine N-methyltransferases , Facteur de nécrose tumorale alpha , Humains , Lignée cellulaire , Inflammation/métabolisme , Inflammation/génétique , Inflammation/anatomopathologie , Interféron gamma/métabolisme , Interféron gamma/pharmacologie , Interleukine-6/métabolisme , Interleukine-6/génétique , Lipopolysaccharides/pharmacologie , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Cadres ouverts de lecture , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Transduction du signal , Facteur de nécrose tumorale alpha/métabolisme , Chromosomes humains de la paire 15
20.
Commun Biol ; 7(1): 753, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902349

RÉSUMÉ

Arginine methylation is catalyzed by protein arginine methyltransferases (PRMTs) and is involved in various cellular processes, including cancer development. PRMT2 expression is increased in several cancer types although its role in acute myeloid leukemia (AML) remains unknown. Here, we investigate the role of PRMT2 in a cohort of patients with AML, PRMT2 knockout AML cell lines as well as a Prmt2 knockout mouse model. In patients, low PRMT2 expressors are enriched for inflammatory signatures, including the NF-κB pathway, and show inferior survival. In keeping with a role for PRMT2 in control of inflammatory signaling, bone marrow-derived macrophages from Prmt2 KO mice display increased pro-inflammatory cytokine signaling upon LPS treatment. In PRMT2-depleted AML cell lines, aberrant inflammatory signaling has been linked to overproduction of IL6, resulting from a deregulation of the NF-κB signaling pathway, therefore leading to hyperactivation of STAT3. Together, these findings identify PRMT2 as a key regulator of inflammation in AML.


Sujet(s)
Inflammation , Leucémie aigüe myéloïde , Souris knockout , Facteur de transcription NF-kappa B , Protein-arginine N-methyltransferases , Transduction du signal , Animaux , Femelle , Humains , Mâle , Souris , Lignée cellulaire tumorale , Inflammation/métabolisme , Inflammation/génétique , Protéines et peptides de signalisation intracellulaire , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Souris de lignée C57BL , Facteur de transcription NF-kappa B/métabolisme , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE