Cell DivisionPub Date : 2025-05-22DOI: 10.1186/s13008-025-00156-y
Fang Liu, Sheng Jin
{"title":"METTL16 accelerates lung adenocarcinoma progression by inducing N6-methyladenosine modification of GTSE1 to regulate p53 pathway and cell cycle.","authors":"Fang Liu, Sheng Jin","doi":"10.1186/s13008-025-00156-y","DOIUrl":"10.1186/s13008-025-00156-y","url":null,"abstract":"<p><strong>Background: </strong>METTL16 has recently emerged as an N6-methyladenosine (m<sup>6</sup>A) methyltransferase that serves an integral role in tumor regulation. However, its involvement in lung adenocarcinoma (LUAD) remains unexamined. This investigation aims to explore METTL16's role and mechanism in LUAD progression.</p><p><strong>Methods: </strong>The expression of METTL16 and G2 and S phase-expressed-1 (GTSE1) in LUAD was evaluated by qRT-PCR or western blotting. LUAD cell malignancy was checked by CCK-8, wound healing, and transwell invasion assays. The relationship among METTL16 and GTSE1 was determined via Pearson correlation analysis and MeRIP assay. The p53 pathway-related proteins were detected by western blotting, and cell cycle was analyzed by flow cytometry.</p><p><strong>Results: </strong>METTL16 was elevated in LUAD, and its silencing significantly reduced LUAD cell proliferation, migration, and invasion. GTSE1 was significantly downregulated upon silencing METTL16. Furthermore, increased levels of GTSE1 mRNA and protein were found in LUAD, and it was correlated positively with METTL16 in LUAD tissues. The stability of GTSE1 was modulated by METTL16 in an m<sup>6</sup>A-dependent way, and GTSE1 overexpression partially rescued the suppressive effects METTL16 silencing on LUAD cells. In addition, GTSE1 overexpression also inhibited p53 pathway to promote LUAD cell cycle.</p><p><strong>Conclusions: </strong>These results indicate that METTL16-mediated m<sup>6</sup>A modification of GTSE1 accelerates LUAD progression by regulating p53 pathway and cell cycle. The aforementioned findings suggest METTL16 and GTSE1 may serve as potential targets for LUAD management.</p>","PeriodicalId":49263,"journal":{"name":"Cell Division","volume":"20 1","pages":"12"},"PeriodicalIF":2.8,"publicationDate":"2025-05-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12096577/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144128960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cell DivisionPub Date : 2025-04-26DOI: 10.1186/s13008-025-00153-1
Otakar Fiser, Petr Muller
{"title":"Role of HSF1 in cell division, tumorigenesis and therapy: a literature review.","authors":"Otakar Fiser, Petr Muller","doi":"10.1186/s13008-025-00153-1","DOIUrl":"https://doi.org/10.1186/s13008-025-00153-1","url":null,"abstract":"<p><p>Heat shock factor 1 (HSF1) is the master orchestrator of the heat shock response (HSR), a critical process for maintaining cellular health and protein homeostasis. These effects are achieved through rapid expression of molecular chaperones, the heat shock proteins (HSPs), which ensure correct protein folding, repair, degradation and stabilization of multiprotein complexes. In addition to its role in the HSR, HSF1 influences the cell cycle, including processes such as S phase progression and regulation of the p53 pathway, highlighting its importance in cellular protein synthesis and division. While HSF1 activity offers neuroprotective benefits in neurodegenerative diseases, its proteome-stabilizing function may also reinforce tumorigenic transformation. HSF1 overexpression in many types of cancer reportedly enhances cell growth enables survival, alters metabolism, weakens immune response and promotes angiogenesis or epithelial-mesenchymal transition (EMT) as these cells enter a form of \"HSF1 addiction\". Furthermore, the client proteins of HSF1-regulated chaperones, particularly Hsp90, include numerous key players in classical tumorigenic pathways. HSF1 thus presents a promising therapeutic target for cancer treatment, potentially in combination with HSP inhibitors to alleviate typical initiation of HSR upon their use.</p>","PeriodicalId":49263,"journal":{"name":"Cell Division","volume":"20 1","pages":"11"},"PeriodicalIF":2.8,"publicationDate":"2025-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12034185/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144058706","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cell DivisionPub Date : 2025-04-23DOI: 10.1186/s13008-025-00155-z
Zhiying Lu, Jing Chen, Chao Luo
{"title":"The m6A modification of LncRNA LINC00200 regulated by WTAP accelerates glioma tumorigenesis by regulating Wnt/β-catenin pathway.","authors":"Zhiying Lu, Jing Chen, Chao Luo","doi":"10.1186/s13008-025-00155-z","DOIUrl":"https://doi.org/10.1186/s13008-025-00155-z","url":null,"abstract":"<p><strong>Background: </strong>Several studies have delineated that dysregulated N6-methyladenosine (m6A) regulators participate in glioma progression. The objective of this study is to investigate the mechanism of Wilms' tumor 1-associating protein (WTAP)-mediated m6A modification of long noncoding RNA (lncRNA) LINC00200 in glioma.</p><p><strong>Methods: </strong>The LINC00200 expression in glioma was analyzed by qRT-PCR. The expressions of WTAP and Wnt/β-catenin pathway associated proteins were determined via qRT-PCR or western blotting. The levels of WTAP-mediated m6A modification of LINC00200 was ascertained by MeRIP-qPCR. Functionally, the effects of LINC00200 knockdown and the interaction of WTAP with LINC00200 on the glioma cell characteristics were examined by CCK8, colony formation, and transwell migration/invasion assays. In vivo experiments were performed to verify the effect of LINC00200 on tumor growth.</p><p><strong>Results: </strong>LINC00200 was overexpressed in glioma, and high LINC00200 level was related to higher-grade tumor. Moreover, its knockdown inhibited the malignant properties and expression of molecules related to Wnt/β-catenin pathway in glioma cell lines. In vivo, LINC00200 knockdown attenuated tumor growth. WTAP was also overexpressed in glioma tissues and demonstrated a positive association with LINC00200 expression. Furthermore, the relative enrichment of LINC00200 m6A was enhanced/reduced in a WTAP-dependent manner. Meanwhile, silencing LINC00200 partially reversed the malignant effects of WTAP overexpression in glioma.</p><p><strong>Conclusion: </strong>These results demonstrate that WTAP-mediated m6A modification of LINC00200 promotes glioma progression by modulating Wnt/β-catenin pathway.</p>","PeriodicalId":49263,"journal":{"name":"Cell Division","volume":"20 1","pages":"10"},"PeriodicalIF":2.8,"publicationDate":"2025-04-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12020130/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144051663","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cell DivisionPub Date : 2025-04-18DOI: 10.1186/s13008-025-00152-2
Liming Cao, Wei Bi
{"title":"METTL16/IGF2BP2 axis enhances malignant progression and DDP resistance through up-regulating COL4A1 by mediating the m6A methylation modification of LAMA4 in hepatocellular carcinoma.","authors":"Liming Cao, Wei Bi","doi":"10.1186/s13008-025-00152-2","DOIUrl":"https://doi.org/10.1186/s13008-025-00152-2","url":null,"abstract":"<p><strong>Background: </strong>Hepatocellular carcinoma (HCC) is the third most common malignant tumor after gastric cancer and esophageal cancer, which is a serious threat to human health. Methyltransferase-like protein 16 (METTL16) regulates the occurrence and development of various cancers, but its molecular mechanism in HCC has not been fully investigated.</p><p><strong>Methods: </strong>A series of databases were used to predict gene expression, methylation sites, correlation analysis, and protein interaction analysis. Gene expression levels were detected by quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and immunohistochemistry (IHC). What's more, drug-resistant cell lines were established for drug resistance analysis. Cell proliferation was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and 5-ethynyl-2'-deoxyuridine (EdU) staining. Flow cytometry, transwell and wound healing assays were used for apoptosis, invasion and migration, respectively. In addition, the regulatory mechanism of METTL16 in HCC was investigated by methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP) and co-immunoprecipitation (Co-IP). Finally, constructing subcutaneous transplanted tumor in nude mice confirmed the effect of METTL16 in vivo.</p><p><strong>Results: </strong>METTL16 was up-regulated in HCC drug-resistant tissues and cells. Knockdown of METTL16 inhibited Cisplatin (DDP) resistance, proliferation, invasion and migration of HCC cells, but promoted apoptosis. Besides, laminin subunit alpha 4 (LAMA4), which was overexpressed in HCC drug-resistant tissues and cells, was selected as the target of METTL16. Mechanistically, METTL16 and m6A reader insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2) co-regulated the m6A modification and mRNA stability of LAMA4, and LAMA4 weakened the effects of METTL16 knockdown on HCC drug-resistance. Meanwhile, LAMA4 bound to collagen type IV alpha 1 chain (COL4A1) and facilitated DDP resistance and HCC progression via COL4A1. Similarly, in vivo, METTL16 induced tumor growth, as well as LAMA4 and COL4A1 expression, and increased DDP resistance.</p><p><strong>Conclusion: </strong>METTL16 and IGF2BP2 jointly mediated the m6A methylation modification of LAMA4, thereby promoting DDP resistance and malignant progression of HCC through regulation of COL4A1.</p>","PeriodicalId":49263,"journal":{"name":"Cell Division","volume":"20 1","pages":"9"},"PeriodicalIF":2.8,"publicationDate":"2025-04-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12008873/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144023195","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"YAP1 activates SLC2A1 transcription and augments the malignant behavior of colorectal cancer cells by activating the Wnt/β-catenin signaling pathway.","authors":"Kunpeng Li, Ya-Jie Dai, Haifeng Zhang, Zhigang Zhang","doi":"10.1186/s13008-025-00148-y","DOIUrl":"10.1186/s13008-025-00148-y","url":null,"abstract":"<p><strong>Objective: </strong>This paper examined the role of solute carrier family 2 member 1 (SLC2A1) in colorectal cancer (CRC) progression, focusing on its expression levels, functional implications, and regulatory mechanisms involving Yes-associated protein 1 (YAP1) and the Wnt signaling pathway.</p><p><strong>Methods: </strong>GEO datasets (GSE14297, GSE18462, GSE40367) were analyzed to identify genes linked to metastasis in CRC, and TCGA-COAD system was used to analyze the expression pattern and prognostic values of SLC2A1 in CRC. Functional studies were conducted using CRC cell lines (Caco-2 and SW480). Cell viability, migration and invasion, and apoptosis were examined using EdU assays, Transwell assays, and flow cytometry. YAP1's regulatory role on SLC2A1 was investigated using ChIP-qPCR and luciferase reporter assays. The Wnt/β-catenin agonist SKL2001 was used for functional rescue experiments.</p><p><strong>Results: </strong>SLC2A1 was upregulated in CRC cells, and its upregulation was associated with tumor metastasis and unfavorable outcomes according to bioinformatics. Knockdown of SLC2A1 resulted in reduced cell viability, decreased migration, and increased apoptosis in Caco-2 and SW480 cells. Additionally, YAP1 was identified as a transcriptional activator of SLC2A1. Knockdown of YAP1 decreased SLC2A1 expression and reduced expression of Wnt target genes, thus suppressing malignant behavior of tumor cells. However, further overexpression of SLC2A1 restored cell viability and migration in YAP1-deficient cells. The YAP1- SLC2A1 axis activated the Wnt/β-catenin by reducing GSK3β activity.</p><p><strong>Conclusion: </strong>SLC2A1 is critical in CRC progression, with YAP1 serving as a key regulator of its expression and function. The YAP1-SLC2A1-Wnt axis represents a potential therapeutic target for CRC, providing insights into metabolic adaptations that support tumor growth and metastasis.</p>","PeriodicalId":49263,"journal":{"name":"Cell Division","volume":"20 1","pages":"8"},"PeriodicalIF":2.8,"publicationDate":"2025-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11969700/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143789108","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cell DivisionPub Date : 2025-03-11DOI: 10.1186/s13008-025-00147-z
Zhenzhen Kong, Ping Zhou, Jiahao Xu, Ying Zhang, Yong Wang
{"title":"RFX2 downregulates RASSF1 expression and YAP phosphorylation through Hippo signaling to promote immune escape in lung adenocarcinoma.","authors":"Zhenzhen Kong, Ping Zhou, Jiahao Xu, Ying Zhang, Yong Wang","doi":"10.1186/s13008-025-00147-z","DOIUrl":"10.1186/s13008-025-00147-z","url":null,"abstract":"<p><strong>Objective: </strong>Regulatory Factor X (RFX) transcription factors have been implicated in different cancers. Ras association domain family (RASSF) has been shown clinical significance in lung cancer. This paper was to investigate the interaction of RFX2 and RASSF1 in lung adenocarcinoma (LUAD).</p><p><strong>Methods: </strong>The transcriptome differences of LUAD patients in GSE32863, GSE43458, and GSE21933 datasets were analyzed. A-549 and NCI-H358 cell lines after overexpression of RFX2 were co-cultured with activated CD8<sup>+</sup> T cells, and the release of IFN-γ, GZMB, PRF1 by CD8<sup>+</sup> T cells, and PD-L1 in the LUAD cells were detected. Cell viability, invasion, and apoptosis were analyzed by CCK-8, Transwell, and TUNEL assays. Dual-luciferase assay and ChIP were conducted to detect the interaction between RFX2 and RASSF1 promoter. An in vivo tumor model was constructed to monitor tumor growth. YAP protein levels and phosphorylation were measured. A-549 and NCI-H358 cells treated with DMSO or PY-60 after RFX2 overexpression were co-cultured with activated CD8<sup>+</sup> T cells.</p><p><strong>Results: </strong>RFX2 was notably downregulated in LUAD. RFX2 overexpression increased infiltrating CD8<sup>+</sup> T cells within transplanted tumors and inhibited immune escape, proliferation, and invasion of LUAD cells. RFX2 was enriched in the RASSF1 promoter, and RFX2 activated RASSF1 transcription by binding to the RASSF1 promoter. RASSF1 knockdown reversed the ability of RFX2 overexpression to inhibit immune escape. RFX2 depletion downregulated RASSF1, which reduced YAP phosphorylation, thus affecting the Hippo pathway to promote the immune escape.</p><p><strong>Conclusion: </strong>RFX2 Loss in LUAD downregulates RASSF1 expression and YAP phosphorylation, thereby promoting immune escape through Hippo signaling.</p>","PeriodicalId":49263,"journal":{"name":"Cell Division","volume":"20 1","pages":"7"},"PeriodicalIF":2.8,"publicationDate":"2025-03-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11895337/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143606423","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The role of MicroRNAs in mesenchymal stem cell differentiation into vascular smooth muscle cells.","authors":"Sakhavat Abolhasani, Yasin Ahmadi, Yavar Rostami, Davood Fattahi","doi":"10.1186/s13008-025-00146-0","DOIUrl":"10.1186/s13008-025-00146-0","url":null,"abstract":"<p><p>MicroRNAs (miRNAs) are small, noncoding RNA molecules that play a vital role in regulating gene expression, especially in the differentiation of mesenchymal stem cells (MSCs) into vascular smooth muscle cells (VSMCs). MSCs hold considerable promise for vascular repair and regenerative medicine, given their ability to differentiate into smooth muscle cells (SMCs) under specific molecular cues. Recent studies have shown that miRNAs, through complex regulatory networks, influence MSC differentiation by targeting essential signaling pathways and modulating the expression of differentiation markers, underscoring the intricate roles of these molecules in cellular development.This review comprehensively examines the functions of various miRNAs in MSC differentiation, focusing on miR-143 and miR-145, which are upregulated by transforming growth factor beta 1 (TGF-β1), a key growth factor in SMC development. These miRNAs enhance differentiation by promoting the expression of SMC markers, including α-smooth muscle actin (α-SMA) and calponin, and by inhibiting factors that preserve MSCs in an undifferentiated state. This review further discusses the roles of miR-503, which supports SMC differentiation through SMAD7 inhibition via the TGF-β pathway, and miR-222-5p, which counteracts differentiation by downregulating ROCK2 and α-SMA. By highlighting these regulatory mechanisms, this review aims to clarify the bidirectional and multifaceted role of miRNAs in VSMC differentiation. This study offers insights into the therapeutic potential of miRNA-mediated MSC differentiation for vascular repair and regeneration, ultimately contributing to improved cardiovascular outcomes.</p>","PeriodicalId":49263,"journal":{"name":"Cell Division","volume":"20 1","pages":"6"},"PeriodicalIF":2.8,"publicationDate":"2025-03-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11887104/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143587706","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cell DivisionPub Date : 2025-02-14DOI: 10.1186/s13008-025-00145-1
Huiye Yang, Yuchan He, Fujun Qu, Jie Zhu, Liyuan Deng, Fang Jiang, Xianyi Wu, Yixuan Chen, Ali Kashif, Xiaotao Wang
{"title":"Maraviroc enhances Bortezomib sensitivity in multiple myeloma by inhibiting M2 macrophage polarization via PI3K/AKT/RhoA signaling pathway in macrophages.","authors":"Huiye Yang, Yuchan He, Fujun Qu, Jie Zhu, Liyuan Deng, Fang Jiang, Xianyi Wu, Yixuan Chen, Ali Kashif, Xiaotao Wang","doi":"10.1186/s13008-025-00145-1","DOIUrl":"10.1186/s13008-025-00145-1","url":null,"abstract":"<p><strong>Background: </strong>Multiple myeloma (MM) is a malignancy where drug resistance often leads to relapse or refractory disease. Chemokine receptor 5 (CCR5) has emerged as a novel therapeutic target. However, the role of CCR5-antagonist Maraviroc (MVC) in M2 macrophage polarization and its potential to enhance Bortezomib sensitivity in MM has not been fully explored.</p><p><strong>Methods: </strong>We used human bone marrow samples, RPMI 8226 cells, and THP-1 monocytes to investigate CCL3/CCR5 axis. ELISA measured CCL3/CCR5 levels. Knockdown/overexpression vectors modulated expression. Cell proliferation, apoptosis, and macrophage polarization were assessed using CCK8, flow cytometry, and transwell assays. QRT-PCR analyzed CCL3 expression, and western blotting examined PI3K/AKT/RhoA signaling. CCR5 was targeted via siRNAs or MVC. NOD/SCID mouse model evaluated CCL3/CCR5 effects on macrophage polarization and MVC's impact on Bortezomib efficacy.</p><p><strong>Results: </strong>CCL3, CCR5, and M2 macrophage markers are upregulated in MM patients, with CCL3/CCR5 expression correlating with M2 macrophage polarization. Myeloma-secreted CCL3 and paracrine CCR5 significantly promoted M2 macrophage polarization by activating PI3K/AKT/RhoA signaling, which in turn enhanced myeloma proliferation, inhibited apoptosis, and reduced Bortezomib sensitivity. MVC inhibited M2 macrophage polarization and improved Bortezomib sensitivity in vitro and xenograft mouse myeloma models.</p><p><strong>Conclusions: </strong>MVC reduced macrophage polarization and enhanced Bortezomib sensitivity in MM cells.</p>","PeriodicalId":49263,"journal":{"name":"Cell Division","volume":"20 1","pages":"5"},"PeriodicalIF":2.8,"publicationDate":"2025-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11829472/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143426559","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cell DivisionPub Date : 2025-01-29DOI: 10.1186/s13008-025-00143-3
Tereza Růžičková, Monika Vlachová, Lukáš Pečinka, Monika Brychtová, Marek Večeřa, Lenka Radová, Simona Ševčíková, Marie Jarošová, Josef Havel, Luděk Pour, Sabina Ševčíková
{"title":"Detection of early relapse in multiple myeloma patients.","authors":"Tereza Růžičková, Monika Vlachová, Lukáš Pečinka, Monika Brychtová, Marek Večeřa, Lenka Radová, Simona Ševčíková, Marie Jarošová, Josef Havel, Luděk Pour, Sabina Ševčíková","doi":"10.1186/s13008-025-00143-3","DOIUrl":"10.1186/s13008-025-00143-3","url":null,"abstract":"<p><strong>Background: </strong>Multiple myeloma (MM) represents the second most common hematological malignancy characterized by the infiltration of the bone marrow by plasma cells that produce monoclonal immunoglobulin. While the quality and length of life of MM patients have significantly increased, MM remains a hard-to-treat disease; almost all patients relapse. As MM is highly heterogenous, patients relapse at different times. It is currently not possible to predict when relapse will occur; numerous studies investigating the dysregulation of non-coding RNA molecules in cancer suggest that microRNAs could be good markers of relapse.</p><p><strong>Results: </strong>Using small RNA sequencing, we profiled microRNA expression in peripheral blood in three groups of MM patients who relapsed at different intervals. In total, 24 microRNAs were significantly dysregulated among analyzed subgroups. Independent validation by RT-qPCR confirmed changed levels of miR-598-3p in MM patients with different times to relapse. At the same time, differences in the mass spectra between groups were identified using matrix-assisted laser desorption/ionization time of flight mass spectrometry. All results were analyzed by machine learning.</p><p><strong>Conclusion: </strong>Mass spectrometry coupled with machine learning shows potential as a reliable, rapid, and cost-effective preliminary screening technique to supplement current diagnostics.</p>","PeriodicalId":49263,"journal":{"name":"Cell Division","volume":"20 1","pages":"4"},"PeriodicalIF":2.8,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11776158/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143068525","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cell DivisionPub Date : 2025-01-28DOI: 10.1186/s13008-024-00139-5
Wen Luo, Qiyu Xiao, Ying Fu
{"title":"ZNF169 promotes thyroid cancer progression via upregulating FBXW10.","authors":"Wen Luo, Qiyu Xiao, Ying Fu","doi":"10.1186/s13008-024-00139-5","DOIUrl":"10.1186/s13008-024-00139-5","url":null,"abstract":"<p><strong>Background: </strong>Zinc finger protein 169 (ZNF169) plays a key role in cancer development. However, the specific role of ZNF169 in the tumorigenesis of thyroid carcinoma (THCA) remains poorly understood.</p><p><strong>Methods: </strong>The expression of ZNF169 was measured using immunohistochemistry, RT-qPCR, and western blot. Cell proliferation was detected using CCK-8 assay and cell colony formation assays, while cell migration was determined by Transwell assay. Flow cytometry was used to detect cell apoptosis and cell cycle distribution. The interaction of ZNF169 and its downstream gene was studied using luciferase assay and CHIP-PCR. Recovery assay in cells and animals were also performed to demonstrate the mechanism.</p><p><strong>Results: </strong>ZNF169 was highly expressed in THCA tissues and cells lines compared with matched adjacent non-cancerous thyroid tissues or normal thyroid epithelial cell. Moreover, thyroid cancer cell proliferation and migration were suppressed following ZNF169 knockdown, while were potentiated by ZNF169 overexpression. ZNF169 also regulate THCA cell apoptosis and cell cycle progression. Mechanically, ZNF169 enhanced the transcription activity and expression of F-box/WD repeat-containing protein 10 (FBXW10) via the binding to its promoter. There was a positive correlation between ZNF169 and FBXW10 in THCA patients. In addition, knockdown of FBXW10 suppressed the proliferation of THCA cells. Recovery assays in vitro and in vivo demonstrated that FBXW10 knockdown reversed the effects of ZNF169 overexpression on THCA cell proliferation and tumor growth.</p><p><strong>Conclusions: </strong>In summary, ZNF169 promotes THCA progression via upregulation of FBXW10, which may provide a novel theoretical basis for the development of clinical therapies for THCA.</p>","PeriodicalId":49263,"journal":{"name":"Cell Division","volume":"20 1","pages":"3"},"PeriodicalIF":2.8,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11773758/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143059657","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}