{"title":"Cytokeratin expression in breast cancer: from mechanisms, progression, diagnosis, and prognosis to therapeutic implications.","authors":"Ensiyeh Bahadoran, Sahar Moghbelinejad, Ghazaleh Mohammadi, Hamid Shahbazmohammadi, Zohreh Abdolvahabi, Manijeh Jalilvand, Isareza Zare, Masood Alaei, Sanaz Keshavarz Shahbaz","doi":"10.1080/23723556.2025.2526230","DOIUrl":"10.1080/23723556.2025.2526230","url":null,"abstract":"<p><strong>Background and aim: </strong>Cytokeratins (CKs) are structural proteins vital to epithelial integrity and play key roles in breast cancer progression. This review explores their expression, functions, and therapeutic potential.</p><p><strong>Methods: </strong>A systematic review was performed using PubMed, Scopus, and Google Scholar. We focused on in vivo, in vitro, and human studies - as well as review articles - published through 1982 that included keywords such as KRT5/13/16/17/18/19/23/80, Cytokeratin 5/13/16/17/18/19/23/80, Keratin 5/13/16/17/18/19/23/80, CK5/13/16/17/18/19/23/80, Cancer, Tumor, Breast cancer, Triple-negative breast cancer, and TNBC. Following title, abstract, and full-text screening of extracted studies, irrelevant articles and duplicates were excluded.</p><p><strong>Results: </strong>CK5 and CK17 are strongly associated with aggressive breast cancer subtypes, particularly triple-negative breast cancer (TNBC), influencing tumor invasiveness and drug resistance. CK18 and CK19 play key roles in estrogen receptor signaling and epithelial stability. Newly identified CKs, CK23 and CK80, show strong correlations with metastasis and poor prognosis. CK-driven pathways, such as the Wnt/β-catenin and EMT pathways, contribute to tumor progression and therapy resistance.</p><p><strong>Conclusion: </strong>CKs are key biomarkers for breast cancer classification, prognosis, and therapy response. Their roles in tumor biology suggest potential for targeted treatment and personalized care to improve outcomes.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":"12 1","pages":"2526230"},"PeriodicalIF":2.6,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12218508/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144555202","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"TRIM22 promotes glioblastoma development by ubiquitinating Bcl-2.","authors":"Jiahao Zhang, Yuning Chen, Gaosong Wang, Hui Huang, Yuankun Liu, Jin Huang, Junfei Shao, Jiantong Jiao, Chao Cheng","doi":"10.1080/23723556.2025.2518679","DOIUrl":"10.1080/23723556.2025.2518679","url":null,"abstract":"<p><p>Glioblastoma (GBM) exhibits elevated TRIM22 expression correlated with tumor progression, as validated in TCGA/GEO databases. The effects of TRIM22 knockdown and overexpression on GBM proliferation were evaluated with cellular assays. TRIM22 was identified as a potential Bcl-2 activator via a ubiquitination microarray. Flow cytometry (FCM) was utilized to investigate cell apoptosis. Additionally, the expression levels of Bcl-2 and proteins associated with Bcl-2 were evaluated using Western blot analysis. The interaction and ubiquitination of TRIM22 and Bcl-2 were analyzed via immunoprecipitation (IP). TRIM22 overexpression is correlated with glioma progression, and TRIM22 deficiency inhibits GBM cell proliferation. FCM revealed that TRIM22 knockdown promotes GBM cell apoptosis. A TRIM22-overexpressing ubiquitination microarray identified TRIM22 as a potential activator of Bcl-2. Western blot analysis revealed that TRIM22 increases the protein expression levels of Bcl-2. Ubiquitination assays revealed that TRIM22 promotes the stability of Bcl-2 via nondegradative ubiquitination. IP experiments indicated that TRIM22 binds to Bcl-2. TRIM22 may significantly impact glioma progression by modulating Bcl-2. Previous studies have shown that knockdown of TRIM22 can enhance the sensitivity of temozolomide treatment, so TRIM22 is expected to become a new target for glioma immunotherapy.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":"12 1","pages":"2518679"},"PeriodicalIF":2.6,"publicationDate":"2025-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12184166/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144477148","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
M Rychla, J Hrabeta, P Jencova, N Podhorska, T Eckschlager
{"title":"The Role of V-ATPase ATP6V0D1 Subunit in Chemoresistance and Ellipticine-Induced Cytoplasmic Vacuolation in Neuroblastoma Cells.","authors":"M Rychla, J Hrabeta, P Jencova, N Podhorska, T Eckschlager","doi":"10.1080/23723556.2025.2518774","DOIUrl":"10.1080/23723556.2025.2518774","url":null,"abstract":"<p><p>Drug resistance remains a major obstacle in neuroblastoma treatment. Lysosomal sequestration, facilitated by the V-ATPase proton pump, is one of the mechanisms of chemoresistance. Overexpression of the ATP6V0D1 subunit of V-ATPase, previously reported in various cancers, was also observed in ellipticine-resistant neuroblastoma cells in our study. Neuroblastoma cells also exhibited increased lysosomal capacity and vacuolation after ellipticine treatment. Knockdown of ATP6V0D1, but not ATP6V1H, enhanced ellipticine sensitivity, suppressed proliferation and migration, decreased lysosomal uptake, and induced G2/M arrest in neuroblastoma cell lines. Notably, inhibiting another V-ATPase subunit, ATP6V1H, had no effect, highlighting the specific role of ATP6V0D1 in drug resistance. Ellipticine-induced vacuolation, identified as endoplasmic reticulum swelling, lacked evidence of paraptosis. ATP6V0D1 knockdown suppressed this phenomenon, whereas ATP6V1H silencing did not. Our findings underscore the importance of ATP6V0D1 in neuroblastoma and suggest potential therapeutic strategies targeting V-ATPase for overcoming drug resistance.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":"12 1","pages":"2518774"},"PeriodicalIF":2.6,"publicationDate":"2025-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12184147/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144477147","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"GSTP1 knockdown induces metabolic changes affecting energy production and lipid balance in pancreatic cancer cells.","authors":"Jenna N Duttenhefner, Katie M Reindl","doi":"10.1080/23723556.2025.2518773","DOIUrl":"10.1080/23723556.2025.2518773","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with limited treatment options, underscoring the need for novel therapeutic targets. Metabolic reprogramming is a hallmark of PDAC, enabling tumor cells to sustain rapid proliferation and survive under nutrient-deprived conditions. While glutathione S-transferase pi 1 (GSTP1) is a known regulator of redox homeostasis in PDAC, its role in metabolic adaptation remains unclear. Here, we show that GSTP1 knockdown disrupts PDAC metabolism, leading to downregulation of key metabolic enzymes (ALDH7A1, CPT1A, SLC2A3, PGM1), ATP depletion, mitochondrial dysfunction, and phospholipid remodeling. Phospholipid remodeling, including an increase in phosphatidylcholine (PC) levels, further suggests a compensatory response to metabolic stress. Importantly, GSTP1 knockdown led to elevated lipid peroxidation, increasing 4-hydroxynonenal (4-HNE) accumulation. Treatment with the antioxidant N-acetyl cysteine (NAC) partially restored metabolic gene expression, reinforcing GSTP1's role in the interplay between redox regulation and metabolism in PDAC. By disrupting multiple metabolic pathways, GSTP1 depletion creates potential therapeutic vulnerabilities that could be targeted through metabolic and oxidative stress-inducing therapies to enhance treatment efficacy.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":"12 1","pages":"2518773"},"PeriodicalIF":2.6,"publicationDate":"2025-06-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12169041/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144310462","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yvonne Wettergren, Peter Rolny, Helena Lindegren, Elisabeth Odin, Victoria Rotter Sopasakis, Simon Keane, Katarina Ejeskär
{"title":"Increased <i>MLH1</i>, <i>MGMT</i>, and <i>p16INK4a</i> methylation levels in colon mucosa potentially useful as early risk marker of colon cancer.","authors":"Yvonne Wettergren, Peter Rolny, Helena Lindegren, Elisabeth Odin, Victoria Rotter Sopasakis, Simon Keane, Katarina Ejeskär","doi":"10.1080/23723556.2025.2503069","DOIUrl":"https://doi.org/10.1080/23723556.2025.2503069","url":null,"abstract":"<p><p>The genes MutL Homolog 1 (<i>MLH1</i>), O6-methylguanine-DNA methyltransferase (<i>MGMT</i>), and cyclin-dependent kinase inhibitor <i>p16INK4a</i> are commonly downregulated by hypermethylation in colorectal cancer. Long interspersed nucleotide element 1 (LINE-1) can be used as marker for global hypomethylation. This study compared <i>MLH1</i>, <i>MGMT</i>, <i>p16INK4a</i>, and LINE-1 methylation with gene expression in colon tumors, matched non-cancerous mucosa, and control mucosa to identify signs of premalignancy. Tissues were obtained from 20 colon cancer patients and 40 controls. CpG site methylation was quantified by pyrosequencing, expression by qPCR, and MSI/<i>KRAS</i> status by fragment analysis and droplet digital PCR. <i>MLH1</i>, <i>MGMT</i>, and <i>p16INK4a</i> methylation was increasingly higher in control mucosa, non-cancerous mucosa, and tumors. <i>MLH1</i> expression was lower in tumors compared to non-cancerous mucosa but higher compared to control mucosa. Tumoral LINE-1 methylation correlated negatively with <i>MLH1</i> (<i>r</i> = -0.51, <i>p</i> = .021) and p16INK4a (<i>r</i> = -0.55, <i>p</i> = .012) methylation, but positively (<i>r</i> = 0.74, <i>p</i> = .0002) with <i>MLH1</i> expression. A <i>p16INK4a</i> SNP (rs3814960 C>T) was associated with methylation, expression, and MSI/<i>KRAS</i> status. Aberrant methylation of tumor suppressor genes in colon mucosa could be an early cancer risk marker. Control mucosa is a more reliable reference than non-cancerous mucosa when identifying premalignant changes. Extended studies will evaluate the possible association between rs3814960 and cancer susceptibility. <b>Trial registration</b>: NCT03072641.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":"12 1","pages":"2503069"},"PeriodicalIF":2.6,"publicationDate":"2025-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12068326/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144052226","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"CHK1 inhibition overcomes gemcitabine resistance in non-small cell lung cancer cell A549.","authors":"Zhi-Yin Ke, Tian Fu, Xue-Chun Wang, Xuan Ma, Hai-Han Yin, Wen-Xuan Wang, Yong-Jun Liu, Ai-Ling Liang","doi":"10.1080/23723556.2025.2488537","DOIUrl":"https://doi.org/10.1080/23723556.2025.2488537","url":null,"abstract":"<p><p>The purpose of the study is mainly to investigate anti proliferation of non-small cell lung cancer A549 cells and its mechanism by inhibition of CHK1 expression combined with gemcitabine. The mRNA and protein levels of genes were analyzed by RT-qPCR and Western blot, respectively. Cell viability was detected by CCK-8 assay and clone formation assay. The detection of the cell cycle was used by Annexin V/7-amino-actinomycin D apoptosis detection kit. Analysis of DNA damage was done by immunofluorescence and alkaline comet assay. The results showed that inhibition of CHK1 and gemcitabine combination significantly reduced the proliferation ability of the two cell lines. We also revealed the degradation of full-length PARP and reduced Bcl-2/Bax ratio on increased apoptosis. Inhibition of CHK1 expression leads to DNA damage, induces phosphorylation of γ-H2AX, and affects the repair of homologous recombination ability through Rad51. Mechanistically, gemcitabine increased phosphorylation-ATR and phosphorylation-CHK1, indicating activation of the DNA repair system and ATR-CHK1-CDC25A pathway. Inhibition of CHK1 resulted in increased synthesis of CDK2/Cyclin A2 and CDK2/Cyclin E1 complexes, and more cells entered the subsequent cell cycle, leading to S phase arrest and mitotic catastrophe. We identified inhibition of CHK1 as a potential treatment for NSCLC and confirmed that inhibition of this kinase could overcome acquired gemcitabine resistance.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":"12 1","pages":"2488537"},"PeriodicalIF":2.6,"publicationDate":"2025-04-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11988257/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144031300","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Deepa Bedi, Mohammed Hassan, Alehegne Yirsaw, Biba Vikas, Pran Datta, Temesgen Samuel
{"title":"The immunopeptidome of colon cancer cells treated with topoisomerase inhibiting drug reveals differential as well as common endogenous protein sampling and display of MHC I-associated peptides.","authors":"Deepa Bedi, Mohammed Hassan, Alehegne Yirsaw, Biba Vikas, Pran Datta, Temesgen Samuel","doi":"10.1080/23723556.2025.2471640","DOIUrl":"10.1080/23723556.2025.2471640","url":null,"abstract":"<p><p>Immunotherapy options for microsatellite stable (MSS) colorectal cancer are currently very limited. The lack of detectably unique or altered immunogens in the tumor microenvironment may be a factor. Radiation and chemotherapy may enhance immunotherapy by increasing cancer cell visibility through Major Histocompatibility Complex I (MHC I) expression. To investigate this, we treated MSS and microsatellite-instable (MSI) colon cancer cells with a topoisomerase inhibitor and analyzed MHC I-associated peptides. Treatment increased peptide numbers by 5% in RKO (MSI) cells and 83% in SW620 (MSS) cells, with 40-50% of peptides being exclusive to treatment. Additionally, clustering analysis revealed a set of peptides with uniquely conserved residues displayed only in treated MSS SW620 cells. Gene Ontology analysis of MHC I-displayed proteins revealed a treatment-induced increase in extracellular vesicle- and nuclear-derived proteins, alongside reduced cytosolic protein sampling. Overall, we present evidence for treatment-inducible differential display of peptides, some of which may affect interactions and functions of immune cells. Given the multitude of factors that modulate the effects of increased MHC I expression and associated peptides, further studies are needed to elucidate the pathophysiological implications of these changes.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":"12 1","pages":"2471640"},"PeriodicalIF":2.6,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11881837/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143574152","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"METTL14-mediated m6A modification upregulates HOXB13 expression to activate NF-κB and exacerbate cervical cancer progression.","authors":"Qian Li, Na Zhao, Xuejing Ding, Jufen Zhao","doi":"10.1080/23723556.2024.2423986","DOIUrl":"https://doi.org/10.1080/23723556.2024.2423986","url":null,"abstract":"<p><p>Cervical cancer (CC) is one of the common malignant tumors in women, and the incidence rate is located in the second place of female tumors. As a major RNA N6-methyladenosine (m6A) methyltransferase, methyltransferase-like 14 (METTL14) is involved in tumor progression by catalyzing methylation modifications in mRNAs. However, the molecular mechanism of METTL14-mediated m6A modification in CC remains not fully revealed. The expression of METTL14 was detected by RT-qPCR and western blot. Cell function was assayed by cell counting kit-8 (CCK-8) assay and flow cytometry analysis. Methylated RNA immunoprecipitation (MeRIP) was used to confirm the relationship between METTL14 and homeobox B13 (HOXB13). In our study, we found that the level of METTL14 was elevated in CC tissues and cells compared with their controls. The inhibition of METTL14 significantly impaired cell proliferation and the epithelial-mesenchymal transition (EMT) process, while also induced apoptosis in HeLa and C33A cells. Furthermore, our findings indicated that homeobox B13 (HOXB13) was a target of METTL14, which positively regulated the expression of HOXB13 in an m6A-dependent manner. Rescue experiments indicated that overexpression of HOXB13 effectively reversed the tumor suppression induced by METTL14 knockdown. Finally, we confirmed that METTL14-modified HOXB13 exerted an oncogenic effect through activation of the nuclear factor kappa B (NF-κB) pathway. In conclusion, our data demonstrated that the m6A modification of HOXB13, mediated by METTL14, facilitated the advancement of CC through targeting the NF-κB pathway, which may be a potential molecular target for the treatment of CC.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":"11 1","pages":"2423986"},"PeriodicalIF":2.6,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11556271/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142629584","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Man Hagiyama, Azusa Yoneshige, Tomoyuki Otani, Akihiro Wada, Fuka Takeuchi, Yuji Shoya, Takao Inoue, Akihiko Ito
{"title":"An antibody-drug conjugate for endometrioid carcinoma based on the expression of cell adhesion molecule 1.","authors":"Man Hagiyama, Azusa Yoneshige, Tomoyuki Otani, Akihiro Wada, Fuka Takeuchi, Yuji Shoya, Takao Inoue, Akihiko Ito","doi":"10.1080/23723556.2024.2399379","DOIUrl":"https://doi.org/10.1080/23723556.2024.2399379","url":null,"abstract":"<p><p>Cell adhesion molecule 1 (CADM1), an immunoglobulin superfamily member, is expressed in endometrial glandular cells highly during the proliferative phase but lowly during the secretory phase. Previously, a CADM1-targeting antibody-drug conjugate (ADC) was generated, in which a humanized anti-CADM1 ectodomain antibody h3E1 was linked with monomethyl auristatin E (h3E1-MMAE ADC). The present study aimed at probing whether this ADC could be useful for the treatment of endometrial neoplasm. Firstly, immunohistochemistry for CADM1 was conducted on proliferative-phase endometrium (<i>n</i> = 13), endometrial hyperplasia (<i>n</i> = 35), and endometrioid carcinoma at various stages (<i>n</i> = 166). CADM1 immunostaining intensity was highest in atypical endometrial hyperplasia and endometrioid carcinoma confined within the endometrium and was decreased stepwise as the carcinoma stage progressed. Next, h3E1-MMAE ADC was examined for its cytotoxicity in vitro using human endometrial adenocarcinoma cell lines expressing CADM1; HEC-1B, HEC-50B, JHUM-3, and OMC-2. The ADC killed these cells in a dose-dependent manner with half maximal inhibitory concentration (IC50) of 12.02 nM for HEC-1B and 2.04 nM for HEC-50B. Collectively, h3E1-MMAE ADC may serve as a noninvasive alternative to simple hysterectomy in the treatment of endometrioid carcinoma confined within the endometrium.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":"11 1","pages":"2399379"},"PeriodicalIF":2.6,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11382700/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142297421","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Shahriar Tarighi, Poonam Kumari, Alejandro Vaquero, Thomas Braun, Alessandro Ianni
{"title":"The SIRT7-nucleolus connection in cancer: ARF enters the fray.","authors":"Shahriar Tarighi, Poonam Kumari, Alejandro Vaquero, Thomas Braun, Alessandro Ianni","doi":"10.1080/23723556.2024.2381287","DOIUrl":"10.1080/23723556.2024.2381287","url":null,"abstract":"<p><p>The nucleolar enzyme sirtuin 7 (SIRT7) promotes cancer progression in certain malignancies, likely in part by controlling ribosome biosynthesis. Recently, we discovered that SIRT7 destabilizes the cyclin dependent kinase inhibitor 2A (CDKN2A, known as ARF) within the nucleolus, aiding cancer progression. We propose that targeting nucleolar SIRT7 offers promise for new anti-cancer therapies.</p>","PeriodicalId":37292,"journal":{"name":"Molecular and Cellular Oncology","volume":"11 1","pages":"2381287"},"PeriodicalIF":2.6,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11259054/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141735282","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}