{"title":"SIRT6 regulates the HIPK2/P53 pathway to reduce oxidative stress and apoptosis to attenuate vancomycin-induced nephrotoxicity","authors":"Xiuying Feng , Yunhui Liu , Lei Su , Luyang Xu","doi":"10.1016/j.mrfmmm.2024.111897","DOIUrl":"10.1016/j.mrfmmm.2024.111897","url":null,"abstract":"<div><div>SIRT6 is known to play a protective role in several kidney diseases; however, its role in vancomycin-induced renal injury remains unclear. This study aims to confirm the role and related mechanisms of SIRT6 in vancomycin-induced renal injury. To develop a kidney damage model, mice were given vancomycin injections for seven days. Additionally, an in vivo transfection with a SIRT6 overexpression plasmid was performed. PCR and Western blot analyses were used to assess the SIRT6 mRNA and protein expression levels in renal tissue. HE staining was performed to evaluate renal tissue damage, while Scr and BUN were measured using specialized kits. Renal tissue apoptotic cells were labeled using a TUNEL kit, and the levels of the antioxidant enzymes SOD and GSH were measured using appropriate kits. Western blot was used to identify HIPK2, p-p53, and p53 protein expression in the renal tissue. The results reveal that SIRT6 is expressed at markedly low levels in renal tissue. Furthermore, mice administered vancomycin exhibited a significant increase in Scr and BUN levels, indicating impaired renal function. Histological examination through HE staining demonstrated considerable damage to the renal tissue of the vancomycin group. Additionally, the renal tissue of the mice in the vancomycin group displayed reduced levels of the antioxidant enzymes SOD and GSH, an increased number of TUNEL-positive cells, and significantly elevated levels of HIPK2 and p-p53 protein expression. Moreover, the mice transfected with SIRT6 exhibited significant improvements in previously described symptoms. These findings imply that the inhibition of HIPK2/p53 by SIRT6 may represent a promising therapeutic strategy for alleviating vancomycin-induced nephrotoxicity.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"830 ","pages":"Article 111897"},"PeriodicalIF":1.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143178151","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sepideh Hassanpour Khodaei , Shahnaz Sabetkam , Zeinab Mazloumi , Khadijeh Dizaji Asl , Ali Rafat
{"title":"Targeting cancer-cell mitochondria using Tigecycline improves radiotherapy response in colorectal cancer cell line","authors":"Sepideh Hassanpour Khodaei , Shahnaz Sabetkam , Zeinab Mazloumi , Khadijeh Dizaji Asl , Ali Rafat","doi":"10.1016/j.mrfmmm.2025.111905","DOIUrl":"10.1016/j.mrfmmm.2025.111905","url":null,"abstract":"<div><h3>Background</h3><div>Colorectal cancer (CRC) is the third most common cancer worldwide and causes more than 50,000 deaths in the United States each year. Due to the limited therapeutic options and poor prognosis in CRC, extensive research and development of novel therapeutic methods is essential. In this regard, the presence of cancer stem cells with unlimited division ability is the main reason for the therapeutic resistance in CRC. Tigecycline is a pharmacological mitochondria inhibitor and blocks mitochondria-related cell proliferation in cancer cells. This study investigated the effects of Tigecycline combined with radiotherapy on CRC cell apoptosis.</div></div><div><h3>Methods</h3><div>Human colorectal cancer cells (HCT-116) were treated with Tigecycline, and cell viability was measured with MTT assay. In the next step, the cells were exposed to radiation using a Siemens Primus 6 MV linear accelerator at radiation dose of 400 cGy. Finally, we evaluated cancer cell apoptosis, caspase-3 activity and apoptotic-related genes expression with AnnexinV/PI, flowcytometry and gene expression, respectively.</div></div><div><h3>Results</h3><div>The MTT assay revealed an IC50 value of 93 μM for Tigecycline after 48 hours. Mitochondria inhibition, at its IC50 value, sensitizes colorectal cancer cells to radiotherapy. Compared to monotherapy, the combination therapy increased the number of apoptotic cells and caspase-3 activity, up-regulated pro-apoptotic genes, and down-regulated anti-apoptotic genes.</div></div><div><h3>Conclusion</h3><div>In conclusion, our data suggests that targeting mitochondria may represent a clinically relevant approach to enhance the sensitivity of colorectal cancer cells to therapy. These findings could provide new insights into cancer therapy and might be used as a novel method to improve the current state of CRC therapy.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"830 ","pages":"Article 111905"},"PeriodicalIF":1.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143826376","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Travis K. Worley , Ayah H. Asal, Lo Cooper, Charmain T. Courcelle, Justin Courcelle
{"title":"The complex development of psoralen-interstrand crosslink resistance in Escherichia coli requires AcrR inactivation, retention of a marbox sequence, and one of three MarA, SoxS, or Rob global regulators","authors":"Travis K. Worley , Ayah H. Asal, Lo Cooper, Charmain T. Courcelle, Justin Courcelle","doi":"10.1016/j.mrfmmm.2025.111898","DOIUrl":"10.1016/j.mrfmmm.2025.111898","url":null,"abstract":"<div><div>Crosslinking agents, such as psoralen and UVA radiation, can be effectively used as antimicrobials and for treating several dysplastic conditions in humans, including some cancers. Yet, both cancer cells and bacteria can become resistant to these compounds, making it important to understand how resistance develops. Recently, several mutants were isolated that developed high levels of resistance to these compounds through upregulation of components of the AcrAB-TolC efflux pump. Here, we characterized these mutants and found that resistance specifically requires inactivating mutations of the <em>acrR</em> transcriptional repressor which also retain the <em>marbox</em> sequence found within this coding region. In addition, the presence of any one of three global regulators, MarA, SoxS, or Rob, is necessary and sufficient to bind to the <em>marbox</em> sequence and activate resistance. Notably, although psoralen is a substrate for the efflux pump, these regulators are not naturally responsive to this stress as neither psoralen, UVA, nor crosslink induction upregulates <em>acrAB</em> expression in the absence of mutation.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"830 ","pages":"Article 111898"},"PeriodicalIF":1.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143178152","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Genomic and molecular landscape of gallbladder cancer elucidating pathogenic mechanisms novel therapeutic targets and clinical implications","authors":"Manishankar Kumar , Arun Kumar , Abhinav Srivastav , Ashok Ghosh , Dhruv Kumar","doi":"10.1016/j.mrfmmm.2024.111896","DOIUrl":"10.1016/j.mrfmmm.2024.111896","url":null,"abstract":"<div><div>Gallbladder cancer (GBC) is an aggressive malignancy with a poor prognosis, often diagnosed at advanced stages due to subtle early symptoms. Recent studies have provided a comprehensive view of GBC's genetic and mutational landscape, uncovering crucial pathways involved in its pathogenesis. Environmental exposures, particularly to heavy metals, have been linked to elevated GBC risk. Established signaling pathways, including hormonal, apoptotic, metabolic, inflammatory, and DNA damage repair pathways, are integral to GBC progression, and evidence points to the involvement of specific germline and somatic mutations in its development. Key mutations in genes such as KRAS, TP53, IDH1/2, ERBB, PIK3CA, MET, MYC, BRAF, MGMT, CDKN2A and p16 have been identified as contributors to tumorigenesis, with additional alterations including chromosomal aberrations and epigenetic modifications. These molecular insights reveal several potential therapeutic targets that could address the limited treatment options for GBC. Promising therapeutic avenues under investigation include immune checkpoint inhibitors, tyrosine kinase inhibitors, tumor necrosis factor-related apoptosis-inducing ligands (TRAIL), and phytochemicals. Numerous clinical trials are assessing the efficacy of these targeted therapies. This review provides a detailed examination of GBC's genetic and mutational underpinnings, highlighting critical pathways and emerging therapeutic strategies. We discuss the implications of germline and somatic mutations for early detection and individualized treatment, aiming to bridge current knowledge gaps. By advancing our understanding of GBC's molecular profile, we hope to enhance diagnostic accuracy and improve treatment outcomes, ultimately paving the way for precision medicine approaches in managing GBC.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"830 ","pages":"Article 111896"},"PeriodicalIF":1.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142928669","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Di Zhang, Xinwu Zhang, Shuai Chang, Yao Zhao, Li Zhang
{"title":"E2F1 activates USP19 to affect the stability of c-Myc to facilitate the progression of hepatocellular carcinoma","authors":"Di Zhang, Xinwu Zhang, Shuai Chang, Yao Zhao, Li Zhang","doi":"10.1016/j.mrfmmm.2025.111902","DOIUrl":"10.1016/j.mrfmmm.2025.111902","url":null,"abstract":"<div><h3>Background</h3><div>Hepatocellular carcinoma (HCC) is the most common malignant tumor worldwide with a high mortality rate. Herein, this study aims to explore the molecular mechanisms of E2F transcription factor 1 (E2F1), ubiquitin specific peptidase 19 (USP19) and c-Myc in regulating HCC progression.</div></div><div><h3>Methods</h3><div>RT-qPCR and western blotting were utilized to assess mRNA and protein expression, respectively. The behavior of cells was examined through Methylthiazolyldiphenyl-tetrazolium bromide (MTT), flow cytometry, transwell, and cell sphere formation assays. Glycolysis-related indicators were detected by kits. The interaction between USP19 and c-Myc was measured by co-immunoprecipitation (Co-IP). Dual-luciferase reporter assay and Chromatin Immunoprecipitation (ChIP) assays were used to assess the binding of E2F1 and USP19 promoter. A mouse xenograft model was established for the purpose of analysis <em>in vivo</em>.</div></div><div><h3>Results</h3><div>High level of c-Myc was observed in HCC tissues and cells. Silencing c-Myc results in the suppression of cell migration, invasion, proliferation, and glycolysis or promotion of apoptosis. USP19 directly bound to c-Myc, and maintained its stability by removing ubiquitination on c-Myc. Overexpression of c-Myc in HCC cells rescued the anti-tumor effect of USP19 deletion. E2F1 promoted USP19 transcription, and increased USP19 expression counteracts the effects of E2F1 depletion on cell behaviors. <em>In vivo</em>, USP19 knockdown controlled HCC growth by modulating c-Myc.</div></div><div><h3>Conclusion</h3><div>E2F1 activated USP19 transcription, thereby stabilizing c-Myc via deubiquitination and accelerating HCC progression.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"830 ","pages":"Article 111902"},"PeriodicalIF":1.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143512011","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"CUEDC1 promotes the growth, migration, epithelial-mesenchymal transition and inhibits apoptosis of hepatocellular carcinoma cells via the TGF-β/Smad signaling pathway","authors":"Angjian Zhou , Fuyu Chen , Zhongchao Chen","doi":"10.1016/j.mrfmmm.2025.111900","DOIUrl":"10.1016/j.mrfmmm.2025.111900","url":null,"abstract":"<div><h3>Purpose</h3><div>To explore the precise molecular by which CUEDC1, a 42 kDa protein containing a CUE domain located on chromosome 17q22, contributes to liver cancer metastasis.</div></div><div><h3>Method</h3><div>CUEDC1 protein expression levels were determined in liver cancer cells using Western blot analysis. The expression of CUEDC1 in these cells was silenced through siRNA transfection Cell viability was assessed using the Cell Counting Kit-8 (CCK-8) assay. Cell invasion and migration capabilities were evaluated using Transwell assays. The expression of transforming growth factor-beta (TGF-β)/ small mother against decapentaplegic (Smad) pathway, N-cadherin, alpha -smooth muscle actin (α-SMA), and E-cadherin was detected using Western blot.</div></div><div><h3>Result</h3><div>CUEDC1 expression was found to be elevated in liver cancer cells. Knockdown of CUEDC1 reduced the expression of TGF-β, p-Smad2, and p-Smad3, key components of the TGF-β/Smad pathway. Additionally, CUEDC1 knockdown significantly decreased cell survival, migration, invasion, and the EMT process in liver cancer cells.</div></div><div><h3>Conclusion</h3><div>CUEDC1 knockdown markedly reduces EMT and liver cancer cell metastasis by suppressing the TGF-β/Smad signaling pathway.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"830 ","pages":"Article 111900"},"PeriodicalIF":1.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143394418","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"In silico identification of multidrug resistance gene (MDR)-targeted transposon miRNAs in human cancer","authors":"Seda Mesci̇","doi":"10.1016/j.mrfmmm.2025.111903","DOIUrl":"10.1016/j.mrfmmm.2025.111903","url":null,"abstract":"<div><div>miRNAs are small RNA molecules that regulate gene expression and play important roles in various biological processes in cells. The discovery of miRNAs is also of great importance in cancer research. miRNAs enable the development of new approaches in cancer treatment by regulating gene expression in cancer cells and have an important place in cancer development, treatment, and diagnosis. Multidrug resistance (MDR) in cancer is associated with the overexpression of ATP-binding cassette (ABC) transporter proteins in cancer cells. MDR contributes to the dysregulation of ABC transporter proteins, and miRNAs mediate MDR in various cancers, resulting in drug resistance. In this study, it was aimed to identify new miRNA sequences in genes associated with multidrug resistance in cancer using <em>in silico</em> method. After obtaining the mature human miRNA sequences in the miRBASE database, BLAST analyses were performed with these sequences for five multidrug resistance genes (ABCB1, ABCC3, ABCC10, ABCC11, ABCG2) known to be associated with cancer. The RNAhybrid tool was used to find the minimum free energy hybridization of gene and miRNA. The target genes of pre-miRNAs and the metabolic pathways in which the target genes play a role were identified with GeneMANIA, SRplot, miRTargetLink programs. Phylogenetic trees of miRNAs belonging to genes were created using the MEGA X software. Secondary structures of pre-miRNA sequences were determined using the RNAfold Web Server program. According to the data obtained from the study, 107 miRNAs associated with multidrug resistance were identified in human cancers. Transmembrane transporter, drug transport and response to drug functions, and metabolic activities of miRNA-related pathways of MDR genes in various cancer types were determined. Multidrug resistance (MDR) in cancer is often associated with overexpression of ABC transporter proteins, which can lead to failure of cancer treatments. Additionally, the relationship of miRNAs with ABC transporter proteins constitutes an important research area to understand the mechanisms of drug resistance and develop new treatment strategies.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"830 ","pages":"Article 111903"},"PeriodicalIF":1.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143591512","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"APOC1, transcriptionally regulated by FOXM1, promotes M2 macrophage polarization and cervical cancer progression","authors":"Qing Chai, Yan Qi, Xiaoyan Nie, Huan Wang","doi":"10.1016/j.mrfmmm.2025.111904","DOIUrl":"10.1016/j.mrfmmm.2025.111904","url":null,"abstract":"<div><h3>Background</h3><div>Cervical cancer (CC) is a common malignant tumor in women. M2 macrophages are associated with tumor growth, metastasis, and immunosuppression. Apolipoprotein C1 (APOC1) has been confirmed as an oncogene in CC. However, the role and mechanism of APOC1 in CC progression and M2 macrophages remain to be elucidated.</div></div><div><h3>Methods</h3><div>The effects of APOC1 on CC cell malignant phenotypes were examined by CCK-8, colony formation, wound healing, and transwell assays <em>in vitro</em> and mice transplant tumor model <em>in vivo</em>. M2 macrophage polarization was assessed by qRT-PCR and flow cytometry assays. The interaction between APOC1 and forkhead box M1 (FOXM1) was determined using chromatin immunoprecipitation (ChIP) and luciferase reporter assays.</div></div><div><h3>Results</h3><div>The expression of APOC1 and FOXM1 was upregulated in CC tissues and cells. Knockdown of APOC1 or FOXM1 resulted in the inhibition of cell proliferation, migration, invasion, and EMT. Moreover, the polarization of M2 macrophages was attenuated when APOC1 or FOXM1 was silenced. Mechanistically, FOXM1 transcriptionally activated APOC1 by binding to its promoter. Furthermore, overexpression of APOC1 reversed the inhibitory effects of FOXM1 knockdown on cell proliferation, metastasis, and M2 macrophage polarization. Additionally, the knockdown of APOC1 reduced tumor growth and M2 macrophage polarization in mice.</div></div><div><h3>Conclusion</h3><div>FOXM1/APOC1 axis is involved in the progression of CC and the regulation of M2 macrophages polarization, bringing new hope to the treatment of CC.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"830 ","pages":"Article 111904"},"PeriodicalIF":1.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143696726","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Cytotoxicity of allyl isothiocyanate and its metabolites in hepatocellular carcinoma HepG2 cells","authors":"Takashi Hashimoto , Shino Nakamura , Takeshi Suzuki , Yuka Hasegawa , Kazuki Kanazawa","doi":"10.1016/j.mrfmmm.2025.111899","DOIUrl":"10.1016/j.mrfmmm.2025.111899","url":null,"abstract":"<div><h3>Objective</h3><div>To investigate the mechanisms by which allyl isothiocyanate (AITC) exerts its anticancer effects, the present study investigated the cytotoxic effects of AITC and its metabolites on hepatocellular carcinoma HepG2 cells.</div></div><div><h3>Methods</h3><div>The AITC metabolites, <em>S</em>-(<em>N</em>-allylthiocarbamoyl)-L-glutathione (AITC-GSH), <em>N</em>-acetyl-<em>S</em>-(<em>N</em>-allylthiocarbamoyl)-L-cysteine (NAC-AITC), <em>S</em>-(<em>N</em>-allylthiocarbamoyl)-L-cysteinylglycine (AITC-Cys-Gly), and <em>S</em>-(<em>N</em>-allylthiocarbamoyl)-L-cysteine (AITC-Cys) were synthesized. HepG2 cells were treated with these compounds and AITC and subjected to a cell cycle analysis, HPLC analysis for intracellular AITC metabolites, and intracellular reactive oxygen species (ROS) analysis.</div></div><div><h3>Results</h3><div>AITC, AITC-GSH, and NAC-AITC significantly induced cell cycle arrest in the G<sub>2</sub>/M phase and subsequently enhanced apoptotic cell death. The AITC metabolites, AITC-Cys-Gly and AITC-Cys did not induce cell cycle arrest. A correlation was observed between the intracellular concentration of AITC-GSH and the percentage of cells under G<sub>2</sub>/M arrest after the treatments with AITC, AITC-GSH, and NAC-AITC. AITC derived from HepG2 cells treated with AITC-GSH and NAC-AITC conjugated with endogenous GSH, resulting in an increase in ROS levels. A treatment with a ROS inhibitor canceled cell cycle arrest.</div></div><div><h3>Conclusion</h3><div>The conjugation of intracellular GSH with AITC decreased free reduced GSH levels and increased intracellular ROS levels in HepG2 cells, resulting in cytotoxicity, including cell cycle arrest and apoptosis.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"830 ","pages":"Article 111899"},"PeriodicalIF":1.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143421098","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Vildan Betul Yenigun , Vasfiye Betul Ucar , Zeynep Betul Sari , Ali Ahmed Azzawri , Yasemin Sena Acar , Muhammed Burak Kaplan , Suleyman Nergiz , Hasan Acar
{"title":"Evaluation of the simultaneous effects of KRAS G12V and LCS6 alterations on the behavior of head and neck squamous cell carcinoma","authors":"Vildan Betul Yenigun , Vasfiye Betul Ucar , Zeynep Betul Sari , Ali Ahmed Azzawri , Yasemin Sena Acar , Muhammed Burak Kaplan , Suleyman Nergiz , Hasan Acar","doi":"10.1016/j.mrfmmm.2024.111895","DOIUrl":"10.1016/j.mrfmmm.2024.111895","url":null,"abstract":"<div><h3>Background</h3><div>Head and neck squamous cell carcinomas are the seventh most common cancer accounting for 90 % of malignant neoplasia of the upper respiratory system. <em>KRAS</em> is a very important oncogene, leading to the suppression of apoptosis, and promoting the pathogenesis and development of tumors. MicroRNAs (miRNAs) are highly conserved, small noncoding RNA molecules aberrantly expressed in various pathologies including regulation of tumor and metastasis-associated genes. Variant (rs61764370) of the let-7 miRNA complementary site of <em>KRAS</em> 3’-untranslated region (KRAS-LCS6) has been shown to disrupt the ability of miRNAs to target genes resulting in differential target mRNA and protein expression.</div></div><div><h3>Methods</h3><div>In this study, the effects of variant complementary site LCS6 of the let-7 miRNA in head and neck cancer were investigated <em>in vitro</em> using laryngeal carcinoma HEp-2 carrying G12V and LCS6 alterations in the <em>KRAS</em> gene. Non-cancer HEK-293 cells were also used as control cells.</div></div><div><h3>Results</h3><div>G12V mutation in the <em>KRAS</em> gene increases invasion capacity and is specifically active on the ERK pathway associated with metastasis. Alteration in the LCS6 region of the <em>KRAS</em> gene did not show additional effects compared to cells only carrying G12V mutation. Our results also showed that the coexistence of G12V and LCS6 alterations is lethal to specific cell types, UM-SCC-17A laryngeal cancer cells in our case.</div></div><div><h3>Conclusions</h3><div>The LCS6 region alteration of the <em>KRAS</em> may play a key role in further cancer progression, and more research is needed to fully understand the mechanisms by which the LCS6 alterations promote cancer progression.</div></div>","PeriodicalId":49790,"journal":{"name":"Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis","volume":"830 ","pages":"Article 111895"},"PeriodicalIF":1.5,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142819981","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}