{"title":"TMC5 as a Marker of Tumor-Associated Telocytes in Hepatocellular Carcinoma.","authors":"Ying Xu, Jing Yu","doi":"10.31083/FBL36583","DOIUrl":"https://doi.org/10.31083/FBL36583","url":null,"abstract":"<p><strong>Background: </strong>Tumor-associated telocytes (TATCs) perform a pivotal role in hepatocellular carcinoma (HCC) progression and correlate with poor patient outcomes. This study aims to identify specific markers of TATCs in HCC using single-nucleus RNA sequencing (snRNA-seq) and transcriptomic analyses.</p><p><strong>Methods: </strong>Comprehensive snRNA-seq and transcriptomic profiling were performed on HCC and adjacent non-cancerous tissues to detect differential expressed genes (DEGs) in TATCs. Bioinformatics tools, including STING and Cytoscape software, were employed to analyze protein-protein interactions and hub genes. Immune cell interactions were assessed via ligand-receptor network analysis.</p><p><strong>Result: </strong>TATCs constituted 0.35% of cells in HCC tissues, with reduced proportions compared to para-cancerous tissues (0.35% vs 8.19%). Hub genes, including <i>TOP2A</i> (DNA topoisomerase Ⅱ alpha), <i>BUB1B</i> (BUB1 mitotic checkpoint serine/threonine kinase B), <i>KIF11</i> (kinesin family member 11), and <i>CENPF</i> (centromere protein F) were identified in telocytes (TCs). Transcriptomics revealed 622 upregulated and 758 downregulated genes in TATCs versus TCs. <i>TMC5</i> (transmembrane channel like 5) and <i>SLC35F3</i> (solute carrier family 35 member F3) emerged as unique TATCs biomarkers, revealing significant associations with poor overall survival (OS) in HCC patients (HR = 1.499 for <i>TMC5</i>; HR = 1.562 for <i>SLC35F3</i>).</p><p><strong>Conclusion: </strong><i>TMC5</i> and <i>SLC35F3</i> are promising biomarkers for TATCs in HCC, warranting further validation to explore their clinical and therapeutic implications.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"30 4","pages":"36583"},"PeriodicalIF":3.3,"publicationDate":"2025-04-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144059999","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"High-LET Proton Irradiation Significantly Alters the Clonogenic and Tumorigenic Potential of Human Breast Cancer Cell Lines <i>In Vitro</i> and <i>In Vivo</i>.","authors":"Margarita Pustovalova, Rita Mohammad, Yuzhe Wang, Wenyu Xue, Philipp Malakhov, Viktor Nekrasov, Elizaveta Kontareva, Zain Nofal, Vyacheslav Saburov, Dmitry Kolesov, Andreyan Osipov, Sergey Leonov","doi":"10.31083/FBL36415","DOIUrl":"https://doi.org/10.31083/FBL36415","url":null,"abstract":"<p><strong>Background: </strong>The implementation of proton beam irradiation (PBI) for breast cancer (BC) treatment is rapidly advancing due to its enhanced target coverage and reduced toxicities to organs at risk. However, the effects of PBI can vary depending on the cell type. This study aimed to explore the effects of PBI on two BC cell lines, MCF7 and MDA-MB-231.</p><p><strong>Methods: </strong>The relative biological effectiveness (RBE) of PBI was assessed using a clonogenic assay. DNA double-strand break (DSB) repair, epithelial-mesenchymal transition (EMT), and filamentous actin (F-actin) were evaluated using immunofluorescence analysis. The extent of entosis and the senescence-associated β-galactosidase (SA-β-gal) activity were estimated by cytochemistry analysis. The influence of the extracellular matrix was evaluated by cultivating cells in both adherent two-dimensional (2D) environments and within 3D fibrin gels of varying stiffness. The metastatic propensity of cells was investigated using migration tests and the cell encapsulation of carboxylate-modified fluorescent nanoparticles. The comparative tumorigenic potential of cells was investigated using an <i>in vivo</i> model of the chick embryo chorioallantoic membrane (CAM).</p><p><strong>Results: </strong>PBI demonstrated superior efficacy in eliminating MCF7 and MDA-MB-231 cells with RBE 1.7 and 1.75, respectively. Following PBI, MDA-MB-231 cells exhibited significantly lower clonogenic survival compared to MCF7, which was accompanied by the accumulation of phosphorylated histone H2AX (γH2AX), p53-binding protein 1 (53BP1) and Rad51 foci of DNA DSBs repair proteins. After surviving 7 days post-PBI, MCF7 cells exhibited 2.5-fold higher levels of the senescence phenotype and entosis compared to the MDA-MB-231 offspring. Both PBI-survived cell lines had greater capability for 2D collective migration, but their metastatic potential was significantly reduced. A significant influence of extracellular matrix stiffness on the correlation between F-actin expression in PBI-survived cells-an indicator of cell stiffness-and their ability to uptake nanoparticles, a trait associated with metastatic potential, was observed. PBI-survived MDA-MB-231RP subline exhibited a hybrid EMT phenotype and a 70% reduction in tumor growth in the <i>in vivo</i> model of the chick embryo CAM. In contrast, PBI-survived MCF7RP cells exhibit mesenchymal-to-epithelial transition (MET)-like features, and their <i>in vivo</i> tumor growth increased by 66% compared to parental cells.</p><p><strong>Conclusions: </strong>PBI triggers various cellular responses in different BC cell lines, influencing tumor growth through mechanisms like DNA damage repair, stress-induced premature senescence (SIPS), and alterations in the stiffness of tumor cell membranes. Our insights into entosis and the effect of extracellular matrix stiffness on metastatic propensity (nanoparticle uptake) enhance the understanding of the role of PBI in","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"30 4","pages":"36415"},"PeriodicalIF":3.3,"publicationDate":"2025-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144063360","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Sesamin Exerts Anti-Tumor Activity in Nasopharyngeal Carcinoma Through Inducing Autophagy and Reactive Oxygen Species Production.","authors":"Deqiang An, Xianyao Jiang, Yucheng Yang","doi":"10.31083/FBL26038","DOIUrl":"https://doi.org/10.31083/FBL26038","url":null,"abstract":"<p><strong>Background: </strong>Sesamin can suppress many cancers, but its effect on nasopharyngeal carcinoma (NPC) is unclear. Herein, we set out to pinpoint the possible changes in NPC due to Sesamin.</p><p><strong>Methods: </strong>The biological function of NPC cells exposed to Sesamin/N-acetyl-L-cysteine (NAC)/3-Methyladenine (3-MA) was detected, followed by evaluation of reactive oxygen species (ROS) production (dichlorodihydrofluorescein diacetate staining) and mitochondrial membrane potential (MMP) (flow cytometry). Proteins pertinent to apoptosis (cleaved caspase-3, cleaved poly (ADP-ribose) polymerase 1 (PARP1)), cell cycle (Cyclin B1), and autophagy (microtubule-associated protein light chain 3 (LC3)-I, LC3-II, Beclin-1, P62) were quantified by Western blot. After the xenografted tumor model in mice was established, the tumor volume and weight were recorded, and Ki-67 and cleaved caspase-3 levels were determined by immunohistochemical analysis.</p><p><strong>Results: </strong>Sesamin inhibited viability, proliferation, cell cycle progression and migration, induced apoptosis, increased ROS production, and decreased MMP in NPC cells. Sesamin elevated cleaved caspase-3/caspase-3, cleaved PARP1/PARP1, and Beclin-1 expressions as well as LC3-II/LC3-I ratio, while diminishing Cyclin B1 and P62 levels. NAC and 3-MA abrogated Sesamin-induced changes as above in NPC cells. Sesamin inhibited the increase of the xenografted tumor volume and weight, down-regulated Ki-67, and up-regulated cleaved caspase-3 in xenografted tumors.</p><p><strong>Conclusion: </strong>Sesamin exerts anti-tumor activity in NPC, as demonstrated by attenuated tumor proliferation and xenografted tumor volume and weight, as well as induced apoptosis in tumor tissues, consequent upon the promotion of autophagy and reactive oxygen species production.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"30 4","pages":"26038"},"PeriodicalIF":3.3,"publicationDate":"2025-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144036868","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Iman Chanchiri, Emil Birch Christensen, Niels Abildgaard, Torben Barington, Thomas Lund, Jakub Krejcik
{"title":"Role of NK Cells in Progression and Treatment of Multiple Myeloma.","authors":"Iman Chanchiri, Emil Birch Christensen, Niels Abildgaard, Torben Barington, Thomas Lund, Jakub Krejcik","doi":"10.31083/FBL26205","DOIUrl":"https://doi.org/10.31083/FBL26205","url":null,"abstract":"<p><p>Multiple myeloma (MM) is a haematological malignancy originating from terminally differentiated B cells, resulting in significant morbidity and mortality. Currently, MM is regarded as an incurable disease, often exhibiting a relapse-remitting pattern that necessitates multiple lines of therapy. It is now well-established that ineffective immunosurveillance plays a critical role in the progression of MM. Consequently, strategies that redirect immune effector cells against MM have emerged as effective treatment modalities, particularly in cases where standard care therapies fail. T cell-based immunotherapy has gained considerable attention in ongoing clinical trials; however, natural killer (NK) cells, known for their ability to execute cytotoxicity against infected and malignant cells with precision, may offer complementary therapeutic advantages over T cells and possess untapped therapeutic potential. This review seeks to introduce readers to the significance of NK cell-mediated immunosurveillance in the context of MM, explore the potential benefits of redirecting NK cells against MM, and illustrate how current treatment strategies are often reliant on the functionality of NK cells. Most importantly, new promising mechanisms of harnessing NK cell-based immunity against MM are reviewed and put into a clinical perspective to highlight their implications for patient treatment and outcomes.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"30 4","pages":"26205"},"PeriodicalIF":3.3,"publicationDate":"2025-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144051599","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lisa Gherardini, Ankush Sharma, Monia Taranta, Caterina Cinti
{"title":"Epigenetic Reprogramming by Decitabine in Retinoblastoma.","authors":"Lisa Gherardini, Ankush Sharma, Monia Taranta, Caterina Cinti","doi":"10.31083/FBL33386","DOIUrl":"https://doi.org/10.31083/FBL33386","url":null,"abstract":"<p><strong>Introduction: </strong>Retinoblastoma (Rb) is a rare cancer, yet it is the most common eye tumor in children. It can occur in either a familial or sporadic form, with the sporadic variant being more prevalent, though its downstream effects on epigenetic markers remain largely unclear. Currently, the treatment for retinoblastoma typically involves aggressive chemotherapy and surgical resection. The identification of specific epigenetic characteristics of non-hereditary (sporadic) Rb has led to the development of advanced, high-throughput methods to explore its epigenetic profile. Our previous research demonstrated that treatment with the demethylating agent 5-Aza-2'-deoxycytidine (decitabine; DAC) induced cell cycle arrest and apoptosis in a well-characterized retinoblastoma model (WERI-Rb-1). Our analysis of time-dependent gene expression in WERI-Rb-1 cells following DAC exposure has led to the development of testable hypotheses to further investigate the epigenetic impact on the initiation and progression of retinoblastoma tumors.</p><p><strong>Methods: </strong>Gene expression analysis of publicly available datasets from patients' primary tumors and normal retina have been compared with those found in WERI-Rb-1 cells to assess the relevance of DAC-driven genes as markers of primary retinoblastoma tumors. The effect of DAC treatment has been evaluated <i>in vivo</i>, both in subcutaneous xenografts and in orthotopic models. qPCR analysis of gene expression and Methylation-Specific PCR (MSP) was performed.</p><p><strong>Results: </strong>Our analysis of network maps for differentially expressed genes in primary tumors compared to DAC-driven genes identified 15 hub/driver genes that may play a pivotal role in the genesis and progression of retinoblastoma. DAC treatment induced significant tumor growth arrest <i>in vivo</i> in both subcutaneous and orthotopic xenograft retinoblastoma models. This was associated with changes in gene expression, either through the direct switching-on of epigenetically locked genes or through the indirect regulation of linked genes, suggesting the potential use of DAC as an epigenetic anti-cancer drug for the treatment of retinoblastoma patients.</p><p><strong>Conclusion: </strong>There is a pressing need to develop innovative treatments for retinoblastoma. Our research revealed that DAC can effectively suppress the growth and progression of retinoblastoma in <i>in vivo</i> models, offering a potential new therapeutic approach to battle this destructive disease. This discovery highlights the impact of this epigenetic therapy in reprogramming tumor dynamics, and thus its potential to preserve both the vision and lives of affected children.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"30 4","pages":"33386"},"PeriodicalIF":3.3,"publicationDate":"2025-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144061238","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"MgATPase Activity is Dispensable for the Pharmacological Regulation of the Functional Effects of the KATP Channels Opening in Brain Mitochondria.","authors":"Olga V Akopova, Anton Smirnov","doi":"10.31083/FBL33450","DOIUrl":"https://doi.org/10.31083/FBL33450","url":null,"abstract":"<p><strong>Background: </strong>The mechanisms underlying the effects of pharmacological mitochondrial ATP-sensitive K<sup>+</sup> channel (mKATP) channel openers on the functional effects of the mKATP channels opening remain disputable. Earlier we have shown that the mKATP channel activation by diazoxide (DZ) occurred at submicromolar concentrations and did not require a MgATP in liver mitochondria. This work aimed to evaluate a requirement of a MgATP for the mKATP channel opening by DZ and its blocking by glibenclamide (Glb) and 5-hydroxy decanoate (5-HD) in rat brain mitochondria and to find the effects of the mKATP channels opening on mitochondrial Ca<sup>2+</sup> uptake, reactive oxygen species (ROS) production, and the mitochondrial permeability transition pore (mPTP).</p><p><strong>Methods: </strong>The mKATP and the mPTP channels activity was assessed by the light scattering; polarography was applied to quantify K<sup>+</sup> transport; Ca<sup>2+</sup> transport and ROS production were monitored with fluorescent probes, chlortetracycline, and dichlorofluorescein, respectively; one-way ANOVA was used for reliability testing.</p><p><strong>Results: </strong>ATP-sensitive K<sup>+</sup> transport in native mitochondria was fully activated by DZ at <0.5 μM and blocked by Glb and 5-HD in the absence of a MgATP, however, Mg<sup>2+</sup> was indispensable for the blockage of the mKATP channel by ATP. DZ increased Ca<sup>2+</sup> uptake, but ROS production was regulated differently: suppressed in mitochondria respiring on glutamate, but activated on succinate. However, in the presence of rotenone, ROS production was suppressed by DZ, which indicated the involvement of reverse electron transport (RET) in the modulation of ROS production. In all cases, the mKATP channel blockers reversed the effects of DZ. The impact of DZ on the mPTP opening strongly correlated with its effects on ROS production. DZ inhibited the mPTP activity on glutamate but elevated on succinate, which was strongly suppressed by rotenone. In the presence of rotenone, the mPTP was strongly inhibited by DZ, which indicated the involvement of ROS and RET in the mechanism of mPTP regulation by DZ.</p><p><strong>Conclusions: </strong>Brain mKATP channel exhibited high sensitivity to DZ on the low sub-micromolar scale; its regulation by DZ and Glb did not require a MgATPase activity; the impact of DZ on the mPTP activity was critically dependent on the regulation of ROS production by ATP-sensitive K<sup>+</sup> transport.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"30 4","pages":"33450"},"PeriodicalIF":3.3,"publicationDate":"2025-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144058597","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Weiwei Dai, Min Zhu, Yujing Sun, Baohong Xu, Guorong Ma, Haiyun Shi, Peng Li
{"title":"NELFCD Promotes Colon Cancer Progression by Regulating the DUSP2-p38 Axis.","authors":"Weiwei Dai, Min Zhu, Yujing Sun, Baohong Xu, Guorong Ma, Haiyun Shi, Peng Li","doi":"10.31083/FBL25221","DOIUrl":"https://doi.org/10.31083/FBL25221","url":null,"abstract":"<p><strong>Background: </strong>To investigate the significance of the negative elongation factor complex member C/D (NELFCD) in colon cancer progression.</p><p><strong>Methods: </strong>Immunohistochemistry staining, Western blot analysis, and real-time quantitative polymerase chain reaction (RT-qPCR) were used to quantify the protein/gene levels. NELFCD-protein arginine methyltransferase 5 (PRMT5) interaction was determined by co-immunoprecipitation assay. A chromatin immunoprecipitation (ChIP) assay was performed to determine the interaction between the promoter region of dual specificity phosphatase 2 (DUSP2), NELFCD, and PRMT5. Cell growth and cell cycle progression were assessed using the cell counting kit-8 proliferation assay, colony formation assay, and/or flow cytometry.</p><p><strong>Results: </strong>NELFCD was upregulated in colon cancer and promoted cancer cell growth. In colon cancer cells, the expression of NELFCD was negatively correlated with DUSP2 expression. The RNA sequencing results indicated that genes in the mitogen-activated protein kinase (MAPK) signaling pathway as well as DUSP2 were affected by NELFCD. The ChIP sequencing results revealed that DUSP2 and genes in the MAPK signaling pathway are direct targets of NELFCD. ChIP assay verified that PRMT5 is enriched at the promoter region of DUSP2 and that NELFCD overexpression promoted this enrichment. A co-immunoprecipitation assay demonstrated that NELFCD was bound to PRMT5, functioning as a macromolecular complex.</p><p><strong>Conclusions: </strong>This study suggests that NELFCD promotes the progression of colon cancer by recruiting PRMT5 to inhibit DUSP2 expression, which subsequently activates the p38 signaling pathway. Targeting the NELFCD-DUSP2-p38 signaling axis may be a promising therapeutic intervention for patients suffering from NELFCD-amplified tumors.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"30 4","pages":"25221"},"PeriodicalIF":3.3,"publicationDate":"2025-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144060746","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Acute Macular Neuroretinopathy Mediated by COVID-19 Infection: Insights into its Clinical Features and Pathogenesis.","authors":"Yixuan Xi, Ziyi Zhou, Tianfang Chang, Guorui Dou, Zhaojie Chu","doi":"10.31083/FBL26412","DOIUrl":"https://doi.org/10.31083/FBL26412","url":null,"abstract":"<p><p>Acute macular neuroretinopathy (AMN) is a rare retinal condition that predominantly affects young females. The incidence of AMN increased significantly during the COVID-19 pandemic, thereby providing a unique opportunity to elucidate the etiology of this disease. In the present study, 24 articles reporting 59 patients were reviewed. The average age of the patients was 33.51 ± 14.02 years, ranging from 16 to 75 years, with females comprising 71.19% of the cases. The average duration of ocular symptoms post-infection was 8.22 ± 10.69 days, ranging from 4 to 150 days. This study investigated the potential pathogenesis of AMN, including the impact of COVID-19 on retinal neurovascular structure and function, immune-mediated inflammatory factor production, blood-retinal barrier disruption, and retinal microvascular damage, as well as potential clinical therapeutic interventions. This research provides a theoretical framework that can inform further investigations of AMN.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"30 4","pages":"26412"},"PeriodicalIF":3.3,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144043994","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fei Qin, Fan Li, Wenxiao Zhao, Suqin Zhang, Jiang Shen, Xinyu Yang
{"title":"M6A Methyltransferase METTL3 Modulates Traumatic Brain Injury by Targeting Ferroptosis.","authors":"Fei Qin, Fan Li, Wenxiao Zhao, Suqin Zhang, Jiang Shen, Xinyu Yang","doi":"10.31083/FBL31304","DOIUrl":"https://doi.org/10.31083/FBL31304","url":null,"abstract":"<p><strong>Background: </strong>Traumatic brain injury (TBI) is a disease caused by external forces that damage brain structure and function. After TBI, iron accumulation and reactive oxygen species (ROS) increase lipid peroxidation, promoting ferroptosis. Methyltransferase-like 3 (<i>METTL3</i>) inhibits ferroptosis by modulating related signaling pathways. This study investigates the effects of <i>METTL3</i> on neuronal ferroptosis in TBI, offering new insights and potential therapies.</p><p><strong>Methods: </strong>TBI mouse and neuron cell models were established and treated with <i>METTL3</i> overexpression. The Morris Water Maze (MWM) test evaluated cognitive function. Histological staining of brain tissues was conducted to assess brain injury, nuclear pyknosis, and iron accumulation. The activation of neurons, microglia, and astrocytes were detected using immunofluorescence staining. Neuron cell proliferation was measured using the Cell Counting Kit 8 (CCK-8). Quantitative PCR (qPCR) and western blot detected the mRNA and protein expression. Ferroptosis was assessed by measuring the accumulation of iron, malondialdehyde (MDA), superoxide dismutase (SOD), and ROS. The quantification of the N6-methyladenosine (m6A) RNA methylation levels in cells was quantified using the m6A-ELISA assay. Methylated RNA immunoprecipitation (MeRIP) assays were conducted to analyze the m6A modification on <i>GPX4</i> mRNA. The interaction between <i>YTHDF2</i> and <i>GPX4</i> mRNA was measured using RNA pulldown and RNA immunoprecipitation (RIP) assays.</p><p><strong>Results: </strong><i>METTL3</i> expression was downregulated in TBI-injured brain tissues. Overexpression of <i>METTL3</i> improved cognitive function and brain recovery while simultaneously reducing ferroptosis and neuroinflammation. <i>METTL3</i> overexpression upregulated <i>GPX4</i> expression both <i>in vitro</i> and <i>in vivo</i>. Further studies indicated that m6A reader protein <i>YTHDF2</i> binds to <i>GPX4</i> mRNA, consequently mediating the <i>METTL3</i>-regulated m6A enrichment and RNA stability of <i>GPX4</i>. Knockdown of <i>GPX4</i> and treatment with ferroptosis inducer abolished the protective effects of <i>METTL3</i> on neurons.</p><p><strong>Conclusion: </strong><i>METTL3</i> exhibits anti-ferroptosis properties and promotes brain injury recovery after TBI by regulating the m6A modification and RNA stability of <i>GPX4</i>.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"30 4","pages":"31304"},"PeriodicalIF":3.3,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144061242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}