9,015 results on '"Tumor suppressor"'
Search Results
2. Prolonged mitosis: A key indicator for detecting stressed and damaged cells
- Author
-
Sparr, Carmen and Meitinger, Franz
- Published
- 2025
- Full Text
- View/download PDF
3. Unveiling the hidden gem: A review of long non-coding RNA NBAT-1 as an emerging tumor suppressor and prognostic biomarker in cancer
- Author
-
Sethi, Subhash Chandra, Singh, Ragini, Sahay, Osheen, Barik, Ganesh Kumar, and Kalita, Bhargab
- Published
- 2025
- Full Text
- View/download PDF
4. Salt-inducible kinases (SIKs) in cancer: mechanisms of action and therapeutic prospects
- Author
-
Manoharan, Ravi
- Published
- 2025
- Full Text
- View/download PDF
5. Use of patient-derived organotypic tumor spheroids for testing of viral vector gene therapy in combination with checkpoint blockade
- Author
-
Domingues, Ana Carolina M., de Oliveira, Soraia B., Tessarollo, Nayara G., Lepique, Ana Paula, Rodrigues, Otavio, Sharova, Tatyana, Lawless, Aleigha, Li, David, Basnet, Madak, Boland, Genevieve M., Cohen, Sonia, Jenkins, Russell W., and Strauss, Bryan E.
- Published
- 2025
- Full Text
- View/download PDF
6. From diagnosis to therapy: The role of LncRNA GAS5 in combatting some cancers affecting women
- Author
-
Hsu, Chou-Yi, Rab, Safia Obaidur, Zwamel, Ahmed Hussein, Oghenemaro, Enwa Felix, Chandra, Muktesh, Rajotiya, Sumit, Hjazi, Ahmed, Prasad, KDV, Atteri, Shikha, and Chauhan, Ashish Singh
- Published
- 2025
- Full Text
- View/download PDF
7. Insights into targeting LKB1 in tumorigenesis
- Author
-
Trelford, Charles B. and Shepherd, Trevor G.
- Published
- 2025
- Full Text
- View/download PDF
8. Molecular insights and clinical implications for the tumor suppressor role of SCFFBXW7 E3 ubiquitin ligase
- Author
-
Qi, Yihang, Rezaeian, Abdol-Hossein, Wang, Jingchao, Huang, Daoyuan, Chen, Hong, Inuzuka, Hiroyuki, and Wei, Wenyi
- Published
- 2024
- Full Text
- View/download PDF
9. Potential function of microRNA miRNA-206 in breast cancer pathogenesis: Mechanistic aspects and clinical implications
- Author
-
Babaeenezhad, Esmaeel, Abdolvahabi, Zohreh, Asgharzadeh, Sahar, Abdollahi, Masume, Shakeri, Sara, Moradi Sarabi, Mostafa, and Yarahmadi, Sahar
- Published
- 2024
- Full Text
- View/download PDF
10. Familial isolated pituitary adenoma is independent of Ahr genotype in a novel mouse model of disease
- Author
-
Shen, Anna L., Moran, Susan M., Glover, Edward N., Lin, Bernice C., Carney, Patrick R., and Bradfield, Christopher A.
- Published
- 2024
- Full Text
- View/download PDF
11. An overview of the functions of p53 and drugs acting either on wild- or mutant-type p53
- Author
-
Huang, Yongmi, Jiao, Zhihao, Fu, Yuqing, Hou, Yue, Sun, Jinxiao, Hu, Feiran, Yu, Shangzhe, Gong, Kexin, Liu, Yiru, and Zhao, Guisen
- Published
- 2024
- Full Text
- View/download PDF
12. Emerging role of miRNA in prostate cancer: A future era of diagnostic and therapeutics
- Author
-
Khan, Mohd Mabood, Sharma, Vineeta, and Serajuddin, Mohammad
- Published
- 2023
- Full Text
- View/download PDF
13. miR-96 and its versatile role in cancer
- Author
-
Bagban, Mohammedazim, Sharma, Khanishka, Saifi, Sana, Ilangovan, Ilamathi, Sultana, Saleema, Numanoğlu, Ekin Nur, and Anshu
- Published
- 2023
- Full Text
- View/download PDF
14. MicroRNA-372 acts as a double-edged sword in human cancers
- Author
-
Tajik, Fatemeh, Alian, Fatemeh, Yousefi, Mohammad, Azadfallah, Ali, Hoseini, Aref, Mohammadi, Forogh, Karimi-Dehkordi, Maryam, and Alizadeh-Fanalou, Shahin
- Published
- 2023
- Full Text
- View/download PDF
15. Combinations of grape seed procyanidin extract and milk thistle silymarin extract against lung cancer — The role of MiR-663a and FHIT
- Author
-
Mao, Jenny T., Xue, Bingye, Lu, Qing-Yi, Lundmark, Laurie, Burns, Windie, Yang, Jieping, Lee, Ru-Po, Glass, Joseph, Qualls, Clifford, and Massie, Larry
- Published
- 2023
- Full Text
- View/download PDF
16. CTLA4 blockade abrogates KEAP1/STK11-related resistance to PD-(L)1 inhibitors.
- Author
-
Skoulidis, Ferdinandos, Araujo, Haniel, Do, Minh, Qian, Yu, Sun, Xin, Cobo, Ana, Le, John, Montesion, Meagan, Palmer, Rachael, Jahchan, Nadine, Juan, Joseph, Min, Chengyin, Yu, Yi, Pan, Xuewen, Arbour, Kathryn, Vokes, Natalie, Schmidt, Stephanie, Molkentine, David, Owen, Dwight, Memmott, Regan, Patil, Pradnya, Marmarelis, Melina, Awad, Mark, Murray, Joseph, Hellyer, Jessica, Gainor, Justin, Dimou, Anastasios, Bestvina, Christine, Shu, Catherine, Riess, Jonathan, Blakely, Collin, Pecot, Chad, Mezquita, Laura, Tabbó, Fabrizio, Scheffler, Matthias, Digumarthy, Subba, Mooradian, Meghan, Sacher, Adrian, Lau, Sally, Saltos, Andreas, Rotow, Julia, Johnson, Rocio, Liu, Corinne, Stewart, Tyler, Goldberg, Sarah, Killam, Jonathan, Walther, Zenta, Schalper, Kurt, Davies, Kurtis, Woodcock, Mark, Anagnostou, Valsamo, Marrone, Kristen, Forde, Patrick, Ricciuti, Biagio, Venkatraman, Deepti, Van Allen, Eliezer, Cummings, Amy, Goldman, Jonathan, Shaish, Hiram, Kier, Melanie, Katz, Sharyn, Aggarwal, Charu, Ni, Ying, Azok, Joseph, Segal, Jeremy, Ritterhouse, Lauren, Neal, Joel, Lacroix, Ludovic, Elamin, Yasir, Negrao, Marcelo, Le, Xiuning, Lam, Vincent, Lewis, Whitney, Kemp, Haley, Carter, Brett, Roth, Jack, Swisher, Stephen, Lee, Richard, Zhou, Teng, Poteete, Alissa, Kong, Yifan, Takehara, Tomohiro, Paula, Alvaro, Parra Cuentas, Edwin, Behrens, Carmen, Wistuba, Ignacio, Zhang, Jianjun, Blumenschein, George, Gay, Carl, Byers, Lauren, Gibbons, Don, Tsao, Anne, Lee, J, Bivona, Trever, Camidge, D, Gray, Jhannelle, Lieghl, Natasha, Levy, Benjamin, Brahmer, Julie, and Garassino, Marina
- Subjects
Animals ,Female ,Humans ,Male ,Mice ,AMP-Activated Protein Kinase Kinases ,Antibodies ,Monoclonal ,B7-H1 Antigen ,Carcinoma ,Non-Small-Cell Lung ,Clinical Trials ,Phase III as Topic ,CTLA-4 Antigen ,Drug Resistance ,Neoplasm ,Immune Checkpoint Inhibitors ,Kelch-Like ECH-Associated Protein 1 ,Lung Neoplasms ,Mutation ,Nitric Oxide Synthase Type II ,T-Lymphocytes ,Tumor Microenvironment ,Tumor Suppressor Proteins ,Genes ,Tumor Suppressor - Abstract
For patients with advanced non-small-cell lung cancer (NSCLC), dual immune checkpoint blockade (ICB) with CTLA4 inhibitors and PD-1 or PD-L1 inhibitors (hereafter, PD-(L)1 inhibitors) is associated with higher rates of anti-tumour activity and immune-related toxicities, when compared with treatment with PD-(L)1 inhibitors alone. However, there are currently no validated biomarkers to identify which patients will benefit from dual ICB1,2. Here we show that patients with NSCLC who have mutations in the STK11 and/or KEAP1 tumour suppressor genes derived clinical benefit from dual ICB with the PD-L1 inhibitor durvalumab and the CTLA4 inhibitor tremelimumab, but not from durvalumab alone, when added to chemotherapy in the randomized phase III POSEIDON trial3. Unbiased genetic screens identified loss of both of these tumour suppressor genes as independent drivers of resistance to PD-(L)1 inhibition, and showed that loss of Keap1 was the strongest genomic predictor of dual ICB efficacy-a finding that was confirmed in several mouse models of Kras-driven NSCLC. In both mouse models and patients, KEAP1 and STK11 alterations were associated with an adverse tumour microenvironment, which was characterized by a preponderance of suppressive myeloid cells and the depletion of CD8+ cytotoxic T cells, but relative sparing of CD4+ effector subsets. Dual ICB potently engaged CD4+ effector cells and reprogrammed the tumour myeloid cell compartment towards inducible nitric oxide synthase (iNOS)-expressing tumoricidal phenotypes that-together with CD4+ and CD8+ T cells-contributed to anti-tumour efficacy. These data support the use of chemo-immunotherapy with dual ICB to mitigate resistance to PD-(L)1 inhibition in patients with NSCLC who have STK11 and/or KEAP1 alterations.
- Published
- 2024
17. Pyrimidine Triones as Potential Activators of p53 Mutants.
- Author
-
Fallatah, Maryam, Demir, Özlem, Law, Fiona, Lauinger, Linda, Baronio, Roberta, Hall, Linda, Bournique, Elodie, Srivastava, Ambuj, Metzen, Landon, Norman, Zane, Buisson, Remi, Amaro, Rommie, and Kaiser, Peter
- Subjects
CETSA ,DSF ,cancer ,mutant p53 ,p53 corrector ,p53 reactivator ,tumor suppressor ,Humans ,Tumor Suppressor Protein p53 ,Pyrimidines ,Cell Proliferation ,Cell Line ,Tumor ,Mutation ,Antineoplastic Agents ,Neoplasms - Abstract
p53 is a crucial tumor suppressor in vertebrates that is frequently mutated in human cancers. Most mutations are missense mutations that render p53 inactive in suppressing tumor initiation and progression. Developing small-molecule drugs to convert mutant p53 into an active, wild-type-like conformation is a significant focus for personalized cancer therapy. Prior research indicates that reactivating p53 suppresses cancer cell proliferation and tumor growth in animal models. Early clinical evidence with a compound selectively targeting p53 mutants with substitutions of tyrosine 220 suggests potential therapeutic benefits of reactivating p53 in patients. This study identifies and examines the UCI-1001 compound series as a potential corrector for several p53 mutations. The findings indicate that UCI-1001 treatment in p53 mutant cancer cell lines inhibits growth and reinstates wild-type p53 activities, including DNA binding, target gene activation, and induction of cell death. Cellular thermal shift assays, conformation-specific immunofluorescence staining, and differential scanning fluorometry suggest that UCI-1001 interacts with and alters the conformation of mutant p53 in cancer cells. These initial results identify pyrimidine trione derivatives of the UCI-1001 series as candidates for p53 corrector drug development.
- Published
- 2024
18. Vimentin is a ubiquitination and degradation substrate of the ubiquitin ligase KPC1
- Author
-
Gottfried, Yossi, Lulu-Shimron, Chen, Goldhirsh, Gilad, Fisher, Yael, Ziv, Tamar, Hoon, Dave S.B., Kravtsova-Ivantsiv, Yelena, and Ciechanover, Aaron
- Published
- 2025
- Full Text
- View/download PDF
19. Chapter 636 - Neurocutaneous Syndromes
- Author
-
Sahin, Mustafa, Ullrich, Nicole, Srivastava, Siddharth, and Pinto, Anna L.
- Published
- 2025
- Full Text
- View/download PDF
20. Identification of PTPN12 Phosphatase as a Novel Negative Regulator of Hippo Pathway Effectors YAP/TAZ in Breast Cancer.
- Author
-
Sarmasti Emami, Sahar, Ge, Anni, Zhang, Derek, Hao, Yawei, Ling, Min, Rubino, Rachel, Nicol, Christopher, Wang, Wenqi, and Yang, Xiaolong
- Subjects
Hippo pathway ,PTPN12 ,TAZ ,YAP ,breast cancer ,cell migration ,cell proliferation ,phosphatase ,tumor suppressor ,Phosphoric Monoester Hydrolases ,Hippo Signaling Pathway ,Genes ,Regulator ,Signal Transduction ,Transcription Factors ,Neoplasms - Abstract
The Hippo pathway plays crucial roles in governing various biological processes during tumorigenesis and metastasis. Within this pathway, upstream signaling stimuli activate a core kinase cascade, involving MST1/2 and LATS1/2, that subsequently phosphorylates and inhibits the transcriptional co-activators YAP and its paralog TAZ. This inhibition modulates the transcriptional regulation of downstream target genes, impacting cell proliferation, migration, and death. Despite the acknowledged significance of protein kinases in the Hippo pathway, the regulatory influence of protein phosphatases remains largely unexplored. In this study, we conducted the first gain-of-functional screen for protein tyrosine phosphatases (PTPs) regulating the Hippo pathway. Utilizing a LATS kinase biosensor (LATS-BS), a YAP/TAZ activity reporter (STBS-Luc), and a comprehensive PTP library, we identified numerous novel PTPs that play regulatory roles in the Hippo pathway. Subsequent experiments validated PTPN12, a master regulator of oncogenic receptor tyrosine kinases (RTKs), as a previously unrecognized negative regulator of the Hippo pathway effectors, oncogenic YAP/TAZ, influencing breast cancer cell proliferation and migration. In summary, our findings offer valuable insights into the roles of PTPs in the Hippo signaling pathway, significantly contributing to our understanding of breast cancer biology and potential therapeutic strategies.
- Published
- 2024
21. Functional test of a naturally occurred tumor modifier gene provides insights to melanoma development.
- Author
-
Garcia-Olazabal, Mateo, Adolfi, Mateus Contar, Wilde, Brigitta, Hufnagel, Anita, Paudel, Rupesh, Lu, Yuan, Meierjohann, Svenja, Rosenthal, Gil G, and Schartl, Manfred
- Abstract
Occurrence of degenerative interactions is thought to serve as a mechanism underlying hybrid unfitness in most animal systems. However, the molecular mechanisms underpinning the genetic interaction and how they contribute to overall hybrid incompatibilities are limited to only a handful of examples. A vertebrate model organism, Xiphophorus , is used to study hybrid dysfunction, and it has been shown from this model that diseases, such as melanoma, can occur in certain interspecies hybrids. Melanoma development is due to hybrid inheritance of an oncogene, xmrk , and loss of a co-evolved tumor modifier. It was recently found that adgre5 , a G protein-coupled receptor involved in cell adhesion, is a tumor regulator gene in naturally hybridizing Xiphophorus species Xiphophorus birchmanni (X. birchmanni) and Xiphophorus malinche (X. malinche). We hypothesized that 1 of the 2 parental alleles of adgre5 is involved in regulation of cell growth, migration, and melanomagenesis. Accordingly, we assessed the function of adgre5 alleles from each parental species of the melanoma-bearing hybrids using in vitro cell growth and migration assays. In addition, we expressed each adgre5 allele with the xmrk oncogene in transgenic medaka. We found that cells transfected with the X. birchmanni adgre5 exhibited decreased growth and migration compared to those with the X. malinche allele. Moreover, X. birchmanni allele of adgre5 completely inhibited melanoma development in xmrk -transgenic medaka, while X. malinche adgre5 expression did not exhibit melanoma suppressive activity in medaka. These findings provide evidence that adgre5 is a natural melanoma suppressor and provide new insight in melanoma etiology. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
22. Ubiquitination Enzymes in Cancer, Cancer Immune Evasion, and Potential Therapeutic Opportunities.
- Author
-
Awan, Aiman B., Osman, Maryiam Jama Ali, and Khan, Omar M.
- Subjects
- *
POST-translational modification , *CYTOLOGY , *CELL cycle , *UBIQUITINATION , *METASTASIS - Abstract
Ubiquitination is cells' second most abundant posttranslational protein modification after phosphorylation. The ubiquitin–proteasome system (UPS) is critical in maintaining essential life processes such as cell cycle control, DNA damage repair, and apoptosis. Mutations in ubiquitination pathway genes are strongly linked to the development and spread of multiple cancers since several of the UPS family members possess oncogenic or tumor suppressor activities. This comprehensive review delves into understanding the ubiquitin code, shedding light on its role in cancer cell biology and immune evasion. Furthermore, we highlighted recent advances in the field for targeting the UPS pathway members for effective therapeutic intervention against human cancers. We also discussed the recent update on small-molecule inhibitors and PROTACs and their progress in preclinical and clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
23. Dual Roles of miR-10a-5p and miR-10b-5p as Tumor Suppressors and Oncogenes in Diverse Cancers.
- Author
-
Singh, Rajan, Ha, Se Eun, Yu, Tae Yang, and Ro, Seungil
- Subjects
- *
TUMOR suppressor genes , *NON-coding RNA , *TUMOR growth , *CANCER invasiveness , *TUMOR microenvironment - Abstract
Cancer is a complex genetic disorder characterized by abnormalities in both coding and regulatory non-coding RNAs. microRNAs (miRNAs) are key regulatory non-coding RNAs that modulate cancer development, functioning as both tumor suppressors and oncogenes. miRNAs play critical roles in cancer progression, influencing key processes such as initiation, promotion, and metastasis. They exert their effects by targeting tumor suppressor genes, thereby facilitating cancer progression, while also inhibiting oncogenes to prevent further disease advancement. The miR-10 family, particularly miR-10a-5p and miR-10b-5p (miR-10a/b-5p), is notably involved in cancer progression. Intriguingly, their functions can differ across different cancers, sometimes promoting and at other times suppressing tumor growth depending on the cancer type and target genes. This review explores the dual roles of miR-10a/b-5p as tumor-suppressive miRNAs (TSmiRs) or oncogenic miRNAs (oncomiRs) in various cancers by examining their molecular and cellular mechanisms and their impact on the tumor microenvironment. Furthermore, we discuss the potential of miR-10a/b-5p as therapeutic targets, emphasizing miRNA-based strategies for cancer treatment. The insights discussed in this review aim to advance our understanding of miR-10a/b-5p's roles in tumor biology and their application in developing innovative cancer therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
24. Tumor Suppressor miR-34a: Potential Biomarker of TACE Response in HCC: J. Zavadil et al.: Tumor Suppressor miR-34a: Potential Biomarker of TACE Response in HCC...
- Author
-
Zavadil, Jan, Juracek, Jaroslav, Cechova, Barbora, Rohan, Tomas, Husty, Jakub, Slaby, Ondrej, Litschmannova, Martina, Uher, Michal, Goldberg, S. Nahum, and Andrasina, Tomas
- Subjects
VASCULAR endothelial growth factors ,CHEMOEMBOLIZATION ,BLOOD plasma ,HEPATOCELLULAR carcinoma ,DISEASE progression - Abstract
Purpose: TACE induces variable systemic effects by producing factors that promote inflammation, oncogenesis, and angiogenesis. Here we compare concentrations of microRNAs (miR-21, miR-210 and miR-34a) and vascular endothelial growth factor (VEGF) in hepatocellular carcinoma (HCC) patients undergoing TACE with degradable (DSM) and nondegradable (DEB) particles and potential use of these biomarker changes for prediction of patient outcomes. Materials and Methods: Overall, 52 patients with HCC treated with DSM TACE (24 patients) and DEB TACE (28 patients) were included in this prospective study. Concentrations of studied biomarkers were measured from blood plasma preprocedurally, immediately (< 90 min) postprocedurally, and 24-h after TACE. Levels were compared between DSM and DEB TACE and correlated with treatment response six and 12 months after the first TACE. Results: Both DSM and DEB TACE elevated plasma levels of miR-21, miR-34a, and miR-210 at 24 h post-procedure compared to baseline levels (FC 1.25–4.0). MiR-34a elevation immediately after TACE was significantly associated with nonprogressive disease compared to those with progressive disease at both six months (FC
a : p = 0.014) and 12 months (FCa : p = 0.029) post-TACE. No significant biomarker changes were found between the embolization particle groups. However, VEGF levels showed a decrease only in the DSM TACE group (FC24 : p = < 0.001). Conclusion: Embolization particle type did not significantly impact miRNA or VEGF changes post-TACE. However, miR-34a elevation immediately after the procedure predicts better patient outcome and may prove useful as a biomarkers for the monitoring of clinical outcomes. Level of Evidence: Level 3 Prospective cohort study. [ABSTRACT FROM AUTHOR]- Published
- 2025
- Full Text
- View/download PDF
25. USP44 regulates HEXIM1 stability to inhibit tumorigenesis and metastasis of oral squamous cell carcinoma.
- Author
-
Chen, Shuai, Wu, Kefan, Zong, Yingrui, Hou, Zhenzhen, Deng, Zhifen, and Xia, Zongping
- Subjects
- *
DEUBIQUITINATING enzymes , *MEDICAL sciences , *LIFE sciences , *CYTOLOGY , *SQUAMOUS cell carcinoma , *LUNGS , *CELL cycle regulation , *PROTEOMICS - Abstract
Oral squamous cell carcinoma (OSCC) is the most frequent type of oral malignancy with high metastasis and poor prognosis. The deubiquitinating enzyme Ubiquitin Specific Peptidase 44 (USP44) regulates the mitotic checkpoint, and its deficiency leads to aneuploidy and increases tumor incidence. However, the role of USP44 in OSCC is not well understood. Herein, we analyzed mRNA sequencing data of OSCC samples downloaded from the TCGA and GEO databases and found that USP44 was decreased in human OSCC tissues and was positively correlated to the survival of OSCC patients. To investigate the biological impact of USP44, we used recombinant lentiviruses to overexpress or knockdown USP44 expression in OSCC cell lines, which were also injected subcutaneously or into the lateral tail vein of Male BALB/c nude mice to model tumorigenesis or lung metastasis in vivo, respectively. The results showed that overexpression of USP44 inhibited malignant cell phenotypes in vitro and suppressed tumor growth and lung metastasis in vivo, while its downregulation had the opposite effects. Comprehensive proteomic analyses through Co-IP mass spectrometry and label-free quantitative LC-MS/MS methods identified 112 differentially expressed proteins positively regulated by USP44, among which 13 were involved in cancer-related pathways including apoptotic signaling and cell cycle regulation. PPI analysis identified Hexamethylene Bis-Acetamide-Inducible Protein 1 (HEXIM1) as the hub protein. Upregulation of USP44 enhanced HEXIM1 protein stability, leading to its higher expression in OSCC cells. Silencing of HEXIM1 further enhanced the malignant phenotype of OSCC cells. At the same time, HEXIM1 knockdown reversed the antitumor effects of USP44. These findings demonstrated that USP44 acted as a critical tumor suppressor in OSCC by inhibiting cell proliferation and metastasis through the stabilization of HEXIM1 protein, suggesting that USP44-HEXIM1 axis is a promising target for OSCC therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Constitutive androstane receptor (CAR) functions as a tumor suppressor via regulating stemness in liver cancer.
- Author
-
Bae, Sarah Da Won, Nguyen, Romario, Yuen, Lawrence, Lam, Vincent, George, Jacob, and Qiao, Liang
- Subjects
- *
CANCER stem cells , *LIVER cancer , *ANDROSTANE receptors , *HEPATOCELLULAR carcinoma , *MEDICAL sciences - Abstract
Constitutive androstane receptor (CAR) is a xenosensor that is almost exclusively expressed in the liver. Studies in rodents suggest an oncogenic role for CAR in liver cancer, but its role in human liver cancer is unclear. We aimed to investigate the functional roles of CAR in human liver cancer with a focus on the liver cancer stem cells. We used bioinformatics to increase our understanding of CAR in human liver cancer and associated stem cell markers. We studied the functional roles of CAR in human liver cancer with a focus on the liver cancer stem cell using siRNA, modulation of CAR activity, and tumorsphere formation assays. We have revealed significant associations between CAR and a wide variety of signalling pathways including stemness signalling. Further in vitro studies have shown that activation of CAR significantly reduces cancer cell stemness and represses proliferation, migration, invasion, and the tumorsphere-forming abilities of liver cancer cells (p < 0.05). Our data demonstrates the unequivocal tumor-suppressive role of CAR in liver cancer. While more detailed mechanistic studies are warranted, the efficacy of CAR xeno-activators in the treatment of advanced hepatocellular carcinoma (HCC) may potentially open a new avenue for liver cancer therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Research trends and hotspots in gastric carcinoma associated exosome: a bibliometric analysis.
- Author
-
Liu, Chunqiu, Guo, Honglei, and Jin, Fangzhou
- Subjects
BIBLIOMETRICS ,STOMACH cancer ,EXOSOMES ,RESEARCH personnel ,CELL death - Abstract
Background: Stomach cancer is considered the fifth most common cancer worldwide. This study utilized bibliometric analysis to construct a visualization map of the relationship between stomach cancer and exosomes, aiming to reveal research trends and emerging themes, and provide direction for future research. Method: Retrieve relevant literature on gastric cancer exosomes in the Web of Science Core Collection (WoSCC) over the past 25 years according to search criteria, and conduct bibliometric and visualization analysis using bibliometric software VOSviewer and CiteSpace. Results: This study included a total of 727 articles, with an overall increasing trend in annual publication output. There were 68 countries involved, with China having the largest number of publications followed by the United States. A total of 957 research institutions were involved, with most of the top 10 institutions in terms of publication output being universities in China. The top 5 journals are Molecular Cancer, Cell death & disease, Cancers, International journal of molecular sciences, and Frontiers in oncology. A total of 4529 authors were involved, with 5 authors having a publication output of no less than 13 articles. A total of 35516 references were cited, with a total number of citations. The top publication is "Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells". Conclusion: Over the past 25 years, researchers have been dedicated to studying the field of exosomes related to gastric cancer, and research in this area is currently progressing steadily. Based on previous studies, exosomes in gastric adenocarcinoma serve as biomarkers, potential therapeutic targets, and post-resistance treatment, which represents current hotspots and emerging frontiers in research. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. miR-198 targets TOPORS : implications for oral squamous cell carcinoma pathogenesis.
- Author
-
Kaushik, Pankhuri, Mishra, Radha, Gopal, Champaka, and Kumar, Arun
- Subjects
TUMOR suppressor genes ,SQUAMOUS cell carcinoma ,TUMOR growth ,INVERSE relationships (Mathematics) ,GENE targeting - Abstract
Background: miRNAs play a critical role in the progression of various diseases, including oral squamous cell carcinoma (OSCC), which represents a major health concern and is one of the leading causes for new cancer cases worldwide. The miRNA dysregulation causes havoc and could be attributed to various factors, with epigenetic silencing of tumor suppressor genes being a major contributor to tumorigenesis. In this study, we have explored the tumor suppressive role of miR-198 in OSCC. Methods: The tumor suppressive effect of miR-198 is established using miRNA analysis in OSCC cell lines, patient samples and xenograft nude mice model. The relationship between the miR-198 and TOPORS is explored using bioinformatics analyses, qRT-PCR, dual-luciferase reporter assay, Western blotting and cancer hall marks assays. The hypermethylation of the MIR198 promoter is confirmed using bisulfite sequencing PCR. Results: We have found miR-198 to be upregulated in OSCC cells treated with 5-Azacytidine, a known DNA methyltransferase inhibitor. Upregulation of miR-198 in 5-Azacytidine treated OSCC cells appears to be due to methylation of the MIR198 promoter. Using bioinformatics analysis and dual-luciferase reporter assay, we have identified TOPORS (TOP1 binding arginine/serine rich protein, E3 ubiquitin ligase) as a novel gene target for miR-198. miR-198-mediated repression of TOPORS decreases cell proliferation and anchorage-independent growth and enhances apoptosis of OSCC cells, which is dependent on the presence of the 3′UTR in TOPORS. An inverse correlation between the expression levels of miR-198 and TOPORS is observed in OSCC patient samples, highlighting the biological relevance of their interaction. Delivery of a synthetic miR-198 mimic to OSCC cells results in a significant decrease in xenograft size in nude mice, potentiating its use in therapeutics. Conclusions: These results suggest that miR-198 is epigenetically silenced in OSCC, which promotes tumor growth, in part, by upregulating the levels of TOPORS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. A missense variant effect map for the human tumor-suppressor protein CHK2.
- Author
-
Gebbia, Marinella, Zimmerman, Daniel, Jiang, Rosanna, Nguyen, Maria, Weile, Jochen, Li, Roujia, Gavac, Michelle, Kishore, Nishka, Sun, Song, Boonen, Rick A., Hamilton, Rayna, Dines, Jennifer N., Wahl, Alexander, Reuter, Jason, Johnson, Britt, Fowler, Douglas M., Couch, Fergus J., van Attikum, Haico, and Roth, Frederick P.
- Subjects
- *
MISSENSE mutation , *PROTEIN kinases , *CHECKPOINT kinase 2 , *SPINDLE apparatus , *SACCHAROMYCES cerevisiae , *TUMOR suppressor proteins , *DNA damage - Abstract
The tumor suppressor CHEK2 encodes the serine/threonine protein kinase CHK2 which, upon DNA damage, is important for pausing the cell cycle, initiating DNA repair, and inducing apoptosis. CHK2 phosphorylation of the tumor suppressor BRCA1 is also important for mitotic spindle assembly and chromosomal stability. Consistent with its cell-cycle checkpoint role, both germline and somatic variants in CHEK2 have been linked to breast and other cancers. Over 90% of clinical germline CHEK2 missense variants are classified as variants of uncertain significance, complicating diagnosis of CHK2-dependent cancer. We therefore sought to test the functional impact of all possible missense variants in CHK2. Using a scalable multiplexed assay based on the ability of human CHK2 to complement DNA sensitivity of Saccharomyces cerevisiae cells lacking the CHEK2 ortholog, RAD53 , we generated a systematic "missense variant effect map" for CHEK2 missense variation. The map reflects known biochemical features of CHK2 while offering new biological insights. It also provides strong evidence toward pathogenicity for some clinical missense variants and supporting evidence toward benignity for others. Overall, this comprehensive missense variant effect map contributes to understanding of both known and yet-to-be-observed CHK2 variants. CHEK2 missense variants have been linked to breast and other cancers; however, ∼90% are classified as variants of uncertain significance. To explore sequence-structure-function relationships and to provide new evidence for clinical variant classification, we used a yeast-complementation assay to test over 8,000 CHEK2 coding variants. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. ARF alters PAF1 complex integrity to selectively repress oncogenic transcription programs upon p53 loss.
- Author
-
Wang, Jinli, Fendler, Nikole L., Shukla, Ashutosh, Wu, Shwu-Yuan, Challa, Ashwini, Lee, Jeon, Joachimiak, Lukasz A., Minna, John D., Chiang, Cheng-Ming, Vos, Seychelle M., and D'Orso, Iván
- Subjects
- *
TRANSCRIPTION factors , *GROWTH differentiation factors , *RNA polymerase II , *BONE morphogenetic proteins , *GENETIC transcription - Abstract
The polymerase associated factor 1 (PAF1) complex (PAF1c) promotes RNA polymerase II (RNA Pol II) transcription at the elongation step; however, how PAF1c transcription activity is selectively regulated during cell fate transitions remains poorly understood. Here, we reveal that the alternative reading frame (ARF) tumor suppressor operates at two levels to restrain PAF1c-dependent oncogenic transcriptional programs upon p53 loss in mouse cells. First, ARF assembles into homo-oligomers to bind the PAF1 subunit to promote PAF1c disassembly, consequently dampening PAF1c interaction with RNA Pol II and PAF1c-dependent transcription. Second, ARF targets the RUNX family transcription factor 1 (RUNX1) to selectively tune gene transcription. Consistently, ARF loss triggers RUNX1- and PAF1c-dependent transcriptional activation of pro-growth ligands (growth differentiation factor/bone morphogenetic protein [GDF/BMP]), promoting a cell-intrinsic GDF/BMP-Smad1/5 axis that aberrantly induce cell growth. Notably, pharmacologic inactivation of GDF/BMP signaling and genetic perturbation of RUNX1 significantly attenuate cell proliferation mediated by dual p53 and ARF loss, offering therapeutic utility. Our data underscore the significance of selective ARF-mediated tumor-suppressive functions through a universal transcriptional regulator. [Display omitted] • ARF binds PAF1 to dismantle PAF1c and dampen PAF1c-dependent transcription in vitro • ARF forms homomers and alters PAF1c integrity in an oligomerization-dependent manner • ARF tunes the expression of PAF1c- and RUNX1-dependent pro-growth genes upon p53 loss • ARF loss elicits PAF1c- and RUNX1-mediated expression of ligands driving cell growth Wang et al. discover that the ARF tumor suppressor, which is induced upon p53 loss, assembles homo-oligomers to dismantle the PAF1c to restrain oncogenic transcription programs. ARF loss elicits PAF1c- and RUNX1-dependent transcriptional activation of select GDF/BMP ligands, ultimately activating a cell-intrinsic, pro-proliferative GDF/BMP-Smad1/5 axis sustaining aberrant cell growth. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. MTSS1: beyond the integration of actin and membrane dynamics.
- Author
-
Matskova, Liudmila, Zheng, Shixing, Kashuba, Elena, Ernberg, Ingemar, and Aspenström, Pontus
- Subjects
- *
SCAFFOLD proteins , *LIFE sciences , *LIPID metabolism , *CYTOLOGY , *EMBRYOLOGY - Abstract
MTSS1 is a ubiquitously expressed intracellular protein known mainly for its involvement in basic cellular processes, such as the regulation of actin organization and membrane architecture. MTSS1 has attracted much attention for its role as a tumor suppressor, being absent or expressed at reduced levels in advanced and metastasizing cancers. Occasionally, MTSS1 is, instead, upregulated in metastasis and, in some cases, even in primary tumors. In addition to these well-established functions of MTSS1 linked to its I-BAR- and WH2-domains, the protein is involved in modulating cell–cell contacts, cell differentiation, lipid metabolism, and vesicle formation and acts as a scaffolding protein for several E3 ubiquitin ligases. MTSS1 is classified as a housekeeping protein and is never mutated despite the several pathologic phenotypes linked to its dysregulation. Despite MTSS1's involvement in fundamental signaling pathways, MTSS1 gene ablation is not ubiquitously lethal, although it affects embryonic development. Due to MTSS1´s involvement in many seemingly disparate processes, with many cases lacking mechanistic explanations, we found it timely to review the recent data on MTSS1's role at the cellular level, as well as in health and disease, to direct further studies on this interesting multifunctional protein. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Epigenetic Modifiers: Exploring the Roles of Histone Methyltransferases and Demethylases in Cancer and Neurodegeneration.
- Author
-
Reed, Lauren, Abraham, Janak, Patel, Shay, and Dhar, Shilpa S.
- Subjects
- *
ALZHEIMER'S disease , *PARKINSON'S disease , *HUNTINGTON disease , *HISTONE methyltransferases , *HISTONE demethylases - Abstract
Simple Summary: Histone methyltransferases (HMTs) and histone demethylases (HDMs) are enzymes that modify histones, the proteins around which DNA is wrapped. These modifications play a crucial role in regulating gene expression by adding or removing methyl groups, which can activate or silence genes. In this review, we explore the roles of HMTs and HDMs in the development of cancer and neurodegenerative diseases, specifically, Alzheimer's, Parkinson's, and Huntington's diseases. Regarding cancer, HMTs and HDMs can drive tumor growth and progression by altering gene expression patterns that regulate cell proliferation and survival. For example, the dysregulation of enzymes like KMT2D (an HMT) and KDM2A (an HDM) is linked to various cancers including gastric cancer by influencing the chromatin structure and gene activity. In neurodegenerative diseases, these enzymes impact the health of neurons by modifying genes involved in brain function. In Alzheimer's, Parkinson's, and Huntington's diseases, changes in HMT and HDM activity can lead to the dysregulation of genes critical for neuron survival, contributing to cognitive decline and motor dysfunction. Herein, we highlight the similarities and differences in how HMTs and HDMs function in cancer and neurodegeneration. Understanding these roles may reveal new therapeutic targets that address the epigenetic underpinnings of both cancer and neurodegenerative diseases, offering hope for innovative treatments. Histone methyltransferases (HMTs) and histone demethylases (HDMs) are critical enzymes that regulate chromatin dynamics and gene expression through the addition and removal of methyl groups on histone proteins. HMTs, such as PRC2 and SETD2, are involved in the trimethylation of histone H3 at lysine 27 and lysine 36, influencing gene silencing and activation. Dysregulation of these enzymes often leads to abnormal gene expression and contributes to tumorigenesis. In contrast, HDMs including KDM7A and KDM2A reverse these methylation marks, and their dysfunction can drive disease progression. In cancer, the aberrant activity of specific HMTs and HDMs can lead to the silencing of tumor suppressor genes or the activation of oncogenes, facilitating tumor progression and resistance to therapy. Conversely, in neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), disruptions in histone methylation dynamics are associated with neuronal loss, altered gene expression, and disease progression. We aimed to comprehend the odd activity of HMTs and HDMs and how they contribute to disease pathogenesis, highlighting their potential as therapeutic targets. By advancing our understanding of these epigenetic regulators, this review provides new insights into their roles in cancer and neurodegenerative diseases, offering a foundation for future research. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Mutational characteristics of extranodal NK/T-cell lymphoma analyzed in relation to clinical prognostic indices.
- Author
-
Lee, Jiyeon, Yoon, Sang Eun, Kim, Seok Jin, Kim, Won Seog, and Cho, Junhun
- Subjects
- *
NUCLEOTIDE sequencing , *TUMOR suppressor genes , *KILLER cells , *SUPPRESSOR mutation , *RAS oncogenes - Abstract
Extranodal NK/T-cell lymphoma (ENKTL) is a malignant lymphoma that is associated with Epstein-Barr virus (EBV) infection and poor prognosis. Several clinical risk stratification tools for ENKTL patients have been developed; however, their relationship with molecular alterations of tumor is unclear. We performed panel-based next generation sequencing (NGS) on formalin-fixed paraffin-embedded tissue of 40 ENKTL patients and analyzed them with the clinicopathological features. Patients with over 60 years of age, non-nasal type, stage III-IV, and distant lymph node involvement were 14 (35.0%), 11 (27.5%), 13 (32.5%), and 11 (27.5%), respectively. EBV DNA was detected in the blood of 30 patients (75.0%). In the NGS analysis, mutations involving the JAK/STAT pathway were the most common (n = 17, 42.5%), followed by epigenetic modifier (n = 12, 30.3%), NF-κB pathway (n = 11, 27.5%), tumor suppressor (n = 10, 25.5%), and RAS/MAPK pathway (n = 9, 22.5%). Among these, alterations involving tumor suppressor (P = 0.022) and RAS/MAPK pathway (P = 0.008) were statistically significant poor prognostic factors. Tumor suppressor gene mutations were statistically significantly related to stage III-IV (P = 0.006), distant lymph node involvement (P = 0.002), and prognostic index for natural killer cell lymphoma-EBV (PINK-E) high risk group (P = 0.017). However, alterations involving RAS/MAPK pathway did not significantly correlated with PINK-E or its components. Alterations involving tumor suppressor genes and RAS/MAPK pathway are associated with poor prognosis in ENKTL patients. Tumor suppressor gene mutations are generally correlated with previously known risk factors; however, RAS/MAPK pathway alterations are not. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. TENT5A mediates the cancer-inhibiting effects of EGR1 by suppressing the protein stability of RPL35 in hepatocellular carcinoma.
- Author
-
Min, Xuejie, Lin, Fen, Zhao, Xinge, Yu, Junming, Ge, Chao, Zhang, Saihua, Li, Xianxian, Zhao, Fangyu, Chen, Taoyang, Tian, Hua, Yan, Mingxia, Li, Jinjun, and Li, Hong
- Abstract
Purpose: Terminal nucleotidyltransferase 5A (TENT5A), recently predicted as a non-canonical poly(A) polymerase, is critically involved in several human disorders including retinitis pigmentosa, cancer and obesity. However, the exact biological role of TENT5A in hepatocellular carcinoma (HCC) has not been elucidated. Methods: The transcription level of TENT5A and clinical correlation were analyzed using the LIRI-JP cohort, the TCGA-LIHC cohort, and clinical tissue samples of HCC patients in our laboratory. Proliferation, migration, and invasion were detected with stably TENT5A overexpressing and knockdown HCC cells in vitro and in vivo. Chromatin immunoprecipitation and dual-luciferase reporter assay were performed to verify the binding of the target protein to DNA. Co-immunoprecipitation and GST pull-down assay combined with mass spectrometry (MS) were used to identify protein interactions. Results: Our study presented here shows that TENT5A is downregulated in HCC tissues, suggesting a shorter overall survival for patients. Gain- and loss-of-function experiments reveal that TENT5A suppresses the proliferation and metastasis, and the residue Gly122 is of great importance to the role of TENT5A in HCC. More importantly, EGR1 (Early growth response 1) directly binds to the TENT5A promoter and promotes TENT5A expression. By interacting with RPL35, TENT5A is involved in ribosome biogenesis and exerts a negative regulatory effect on the mTOR pathway. Conclusions: Our findings illustrate the role of the oncosuppressive function of TENT5A in HCC and suggest that the EGR1/TENT5A/RPL35 regulatory axis may be a promising target for therapeutic strategies in HCC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. Modulation of FOXP3 Gene Expression in OVCAR3 Cells Following Rosmarinic Acid and Doxorubicin Exposure.
- Author
-
Toprak, Veysel, Özdemir, İlhan, Öztürk, Şamil, Yanar, Orhan, Kizildemir, Yusuf Ziya, and Tuncer, Mehmet Cudi
- Subjects
- *
ROSMARINIC acid , *CANCER cell proliferation , *CELL migration , *GENE expression , *INHIBITION of cellular proliferation - Abstract
Background/Objectives: Ovarian cancer has the highest mortality rate in the world. Treatment methods are listed as surgery, chemotherapy, and radiotherapy, depending on the stage of cancer, but developing resistance to chemotherapy increases the need for alternative agents that act on the same pathways. The effects of rosmarinic acid (RA) and doxorubicin (DX) on the activation of FOXP3, an important tumor suppressor gene, in OVCAR3 cells were examined. Materials and Methods: In this study, a human ovarian adenocarcinoma cell line was used. MTT analysis was performed to reveal the result of RA and DX on ovarian cancer cell proliferation. Expression levels of FOXP3 for cell proliferation and Capase-3 for apoptosis were determined by RT-qPCR. The wound healing model was applied to determine cell migration rates. The results were evaluated with one-way ANOVA in an SPSS 20.0 program as p ≤ 0.05. Results: It was determined that RA and DX alone and in combination inhibited the proliferation of OVCAR3 cells in different doses for 24, 48, and 72 h, and caused the cells to die by causing them to undergo apoptosis. Caspase-3 expression increased approximately tenfold in OVCAR3 cells, while FOXP3 expression was upregulated only in RA treatment and was downregulated in DX and RA + DX treatments. Conclusions: According to the results of our study, it was determined that the FOXP3 signaling pathway related to apoptosis, and proliferation was affected by the combination treatment of RA and DX in the OVCAR3 cancer cell line. This shows that RA will gain an important place in cancer treatment with more comprehensive study. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Unraveling the Guardian: p53's Multifaceted Role in the DNA Damage Response and Tumor Treatment Strategies.
- Author
-
Zhang, Han, Xu, Jianxiong, Long, Yuxuan, Maimaitijiang, Ayitila, Su, Zhengding, Li, Wenfang, and Li, Jinyao
- Subjects
- *
EXCISION repair , *NEGATIVE regulatory factor , *TRANSCRIPTION factors , *P53 antioncogene , *POST-translational modification , *TUMOR suppressor genes , *DNA repair - Abstract
DNA damage can lead to mutations that can alter the function of oncogenes or tumor suppressor genes, thus promoting the development of cancer. p53 plays a multifaceted and complex role in the DNA damage response and cancer progression and is known as the 'guardian of the gene'. When DNA damage occurs, p53 is activated through a series of post-translational modifications, which stabilize the protein and enhance its function as a transcription factor. It regulates processes including cell cycle checkpoints, DNA repair and apoptosis, thereby preventing the spread of damaged DNA and maintaining genome integrity. On the one hand, p53 can initiate cell cycle arrest and induce cells to enter the G1/S and G2/M checkpoints, preventing cells with damaged DNA from continuing to proliferate and gaining time for DNA repair. At the same time, p53 can promote the activation of DNA repair pathways, including base excision repair, nucleotide excision repair and other repair pathways, to ensure the integrity of genetic material. If the damage is too severe to repair, p53 will trigger the apoptosis process to eliminate potential cancer risks in time. p53 also plays a pivotal role in cancer progression. Mutations in the p53 gene are frequently found in many cancers, and the mutated p53 not only loses its normal tumor suppressor function but may even acquire pro-cancer activity. Therefore, we also discuss therapeutic strategies targeting the p53 pathway, such as the use of small-molecule drugs to restore the function of wild-type p53, the inhibition of negative regulatory factors and synthetic lethality approaches for p53-deficient tumors. This review therefore highlights the important role of p53 in maintaining genomic stability and its potential in therapeutic strategies for cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Role of MARCH E3 ubiquitin ligases in cancer development.
- Author
-
Behera, Abhayananda, Sachan, Deepanshi, Barik, Ganesh Kumar, and Reddy, Aramati Bindu Madhava
- Abstract
Membrane-associated RING-CH (MARCH) E3 ubiquitin ligases, a family of RING-type E3 ubiquitin ligases, have garnered increased attention for their indispensable roles in immune regulation, inflammation, mitochondrial dynamics, and lipid metabolism. The MARCH E3 ligase family consists of eleven distinct members, and the dysregulation of many of these members has been documented in several human malignancies. Over the past two decades, extensive research has revealed that MARCH E3 ligases play pivotal roles in cancer progression by ubiquitinating key oncogenes and tumor suppressors and orchestrating various signaling pathways. Some MARCH E3s act as oncogenes, while others act as tumor suppressors, and the majority of MARCH E3s play both oncogenic and tumor suppressive roles in a context-dependent manner. Notably, there is special emphasis on the sole mitochondrial MARCH E3 ligase MARCH5, which regulates mitochondrial homeostasis within cancer cells. In this review, we delve into the diverse functions of MARCH E3 ligases across different cancer types, shedding light on the underlying molecular mechanisms mediating their effects, their regulatory effects on cancer and their potential as therapeutic targets. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. A Comprehensive Bioinformatic Analysis Identifies a Tumor Suppressor Landscape of the MEG3 lncRNA in Breast Cancer.
- Author
-
Ahmadi, Alireza, Rezaei, Abedeh, Khalaj-Kondori, Mohammad, and Khajehdehi, Mina
- Abstract
Breast cancer (BC) is the leading cause of cancer mortality in women and a major risk to world health. Therefore, effective strategies are required for prompt diagnosis and treatment. Nowadays, non-coding RNAs (ncRNAs), particularly long ncRNAs (lncRNAs), have assumed a significant role in the prognosis and diagnosis of diseases, including cancer. In the present study, surveying the bioinformatic tools, including the lncRNADisease v2.0, OncoDB, InteractiVenn, GEPIA, RAID, COXPRESdb, DAVID v6.8, GEO2R, and LncSEA, we proposed the Maternally Expressed Gene (MEG3) as a potential biomarker in BC. This lncRNA significantly downregulates in BC and is associated with tumor size, metastasis, and pathological stage. MEG3 expression is downregulated in several types of primary human cancers and tumor cell lines, which raises the possibility that it could act as a tumor suppressor. The results suggest that MEG3 may play a crucial role in fundamental pathways, including apoptosis, and interact with essential genes and proteins such as P53. It may also be associated with the prognosis, proliferation, migration, invasion, and metastasis of BC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
39. TRIM-endous functional network of tripartite motif 29 (TRIM29) in cancer progression and beyond.
- Author
-
Wu, Qitong, Nandi, Deeptashree, and Sharma, Dipali
- Abstract
While most Tripartite motif (TRIM) family proteins are E3 ubiquitin ligases, some members have functions beyond the regulation of ubiquitination, impacting normal physiological processes and disease progression. TRIM29, an important member of the TRIM family, exerts a predominant influence on cancer growth, epithelial-to-mesenchymal transition, stemness and metastatic progression by directly potentiating multiple canonical oncogenic pathways. The cancer-promoting effect of TRIM29 is also evident in metabolic interventions and interference with the efficacy of cancer therapeutics. As expected for any key node in cancer, the expression of TRIM29 is tightly regulated by non-coding RNAs, epigenetic modulation, and post-translational regulation. A systematic discussion of how TRIM29 is regulated in cancer, its influences on cancer progression, and its impact on cancer therapeutics is presented in this review. We also explore the context-dependent alterations between TRIM29 function from oncogenic to tumor suppression. As TRIM29 is involved in multiple aspects of cancer progression, a better understanding of its biological impact in cancer may help improve prognosis and develop novel therapeutic combinations, leading to improved personalized cancer care. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
40. Promoter hypermethylation-mediated downregulation of PAX6 promotes tumor growth and metastasis during the progression of liver cancer
- Author
-
Ching-Hua Yeh, Rou-Yu Chen, Ti-Hui Wu, Shan-Yueh Chang, Tsai-Yuan Hsieh, Yu-Lueng Shih, and Ya-Wen Lin
- Subjects
Hepatocellular carcinoma ,PAX6 ,Tumor suppressor ,CDH1 ,THBS1 ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background The progression of liver cancer is a complicated process that involves genetic and epigenetic changes. Paired box 6 (PAX6) is a critical transcription factor for embryonic development. PAX6 is abnormally methylated in human cancer. The role of the PAX6 gene in the pathogenesis of hepatocellular carcinoma (HCC) is still unclear. Methods Transcriptional silencing of PAX6 mediated by promoter methylation was confirmed using quantitative methylation-specific polymerase chain reaction (PCR) and reverse-transcription (RT)-PCR. Then we conducted gain-and-loss of function approaches to evaluate the function of PAX6 in HCC progression in vitro. Moreover, we designed xenograft mouse models to assess the effect of PAX6 on tumor growth and metastasis. Finally, we used RNA sequencing (RNA-seq) strategy and phenotypic rescue experiments to identify potential targets of PAX6 performing tumor-suppressive function. Results Constitutive expression of PAX6 suppressed anchorage-independent growth and cell invasion in vitro as well as tumor growth and metastasis in xenograft mouse models. In contrast, the inhibition of PAX6 using knockout and knockdown strategies increased tumor growth both in vitro and in vivo. Downregulation of PAX6 by doxycycline depletion partially reversed the malignant phenotypes of HCC cells induced by PAX6. Moreover, we identified E-cadherin (CDH1) and thrombospondin-1 (THBS1) as targets of PAX6. Ultimately, we demonstrated that the knockdown of CDH1 and overexpression of THBS1 in PAX6-expressing HCC cells partly reversed the tumor-suppressive effect. Conclusion PAX6 functions as a tumor suppressor partly through upregulation of CDH1 and downregulation of THBS1. Promoter hypermethylation-mediated suppression of PAX6 reduces the tumor suppressor function in the progression of liver cancer.
- Published
- 2024
- Full Text
- View/download PDF
41. Role of early growth response-1 as a tumor suppressor in oral squamous cell carcinoma
- Author
-
Yudai Shimojukkoku, Phuong Thao Nguyen, Kiyohide Ishihata, Takayuki Ishida, Yuka Kajiya, Yasunobu Oku, Koshiro Kawaguchi, Takahiro Tsuchiyama, Hideto Saijo, Kaori Shima, and Tomonori Sasahira
- Subjects
EGR1 ,Oral cancer ,Tumor suppressor ,Growth ,Migration ,Invasion ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Oral squamous cell carcinoma (OSCC) exhibits pronounced local invasiveness and a propensity for lymph node metastasis. Given its frequent detection at advanced stages and the consequential postoperative functional impairments, the identification of effective molecular markers for early detection and treatment is imperative. Early growth response-1 (EGR-1) serves as a versatile transcription factor expressed across various cell types. Its role in cancer is contentious, acting as either an oncogene or a tumor suppressor gene. Methods This study undertook comprehensive analyses, including big data scrutiny, expression profiling using 50 OSCC samples, and in vitro functional assessments, to elucidate EGR-1’s involvement in OSCC. Results Comparative analysis revealed significantly reduced EGR-1 expression in oral cancer tissues compared to healthy controls or normal oral mucosa. In vitro experimentation with multiple OSCC cell lines demonstrated that EGR-1 curbed cell proliferation, migration, and invasion capabilities. Additionally, it was observed that EGR-1 prompted G0/G1 arrest in OSCC cells by modulating the activity of cell cycle regulators. Conclusions These findings strongly support EGR-1’s tumor-suppressive role in OSCC and hint at the potential for novel OSCC therapies aimed at restoring aberrant EGR-1 function.
- Published
- 2024
- Full Text
- View/download PDF
42. Gremlin-2 is a novel tumor suppressor that negatively regulates ID1 in breast cancer
- Author
-
Jiwoo Jung, Na Hui Kim, Jayeon Park, Dayeon Lim, Minji Kwon, World Gil, Suyeon Jung, Minjeong Go, Chaeeon Kim, Ye Hwang Cheong, Mee-Hyun Lee, Hee Sun Park, Yong-Bin Eom, and Sin-Aye Park
- Subjects
Gremlin-2 ,Breast cancer ,Hypermethylation ,RNA sequencing ,Inhibitor of DNA-binding-1 ,Tumor suppressor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Breast cancer is one of the most common cancers in women and is closely associated with obesity. Gremlin-2 (GREM2), an antagonist for bone morphogenetic proteins (BMPs), has been considered an inhibitor of adipogenic differentiation in adipose-derived stromal/stem cells. However, the role of GREM2 in breast cancer cells remains largely unknown, and its signaling mechanism has yet to be clarified. Methods Bioinformatics analysis was conducted using public databases. Breast cancer cells overexpressing mock or GREM2 were used for in vitro and in vivo studies. Cell viability, colony formation, migration, and animal studies were performed to investigate the role of GREM2 in breast cancer cells. Screening of target genes affected by GREM2 overexpression in breast cancer cells was performed through RNA sequencing (RNA-seq) analysis. Results The expression level of GREM2 mRNA was significantly reduced in both breast cancer tissues and cell lines. Kaplan-Meier analysis showed that low expression of GREM2 and high methylation of the GREM2 promoter were each associated with poor patient survival. The low mRNA expression of GREM2 in breast cancer cells was increased by the demethylating agent decitabine. Breast cancer cells overexpressing GREM2 decreased cell proliferation when compared to control cells, both in vitro and in vivo. Through comparison of RNA-seq analysis between cell lines and tissue samples, gene ontologies that were consistently upregulated or downregulated by GREM2 in breast cancer were identified. In particular, the expression of inhibitor of DNA-binding-1 (ID1) was repressed by GREM2. BMP2 is one of the upstream regulators that increases the expression of ID1, and the expression of ID1 reduced by GREM2 was restored by overexpression of BMP2. Also, the migration ability of breast cancer cells, which had been suppressed by GREM2, was restored by BMP2 or ID1. Conclusions Low expression of GREM2 in breast cancer cells is associated with hypermethylation of the GREM2 promoter, which may ultimately contribute to poor patient survival. GREM2 participates in regulating the expression of various genes, including ID1, and is involved in suppressing the proliferation of breast cancer cells. This suggests that GREM2 has the potential to act as a novel tumor suppressor in breast cancer.
- Published
- 2024
- Full Text
- View/download PDF
43. From regulation to deregulation of p53 in hematologic malignancies: implications for diagnosis, prognosis and therapy
- Author
-
Seyed Esmaeil Ahmadi, Elahe Rahimian, Samira Rahimi, Bahman Zarandi, Mehran Bahraini, Maral Soleymani, Seyed Mehrab Safdari, Ashkan Shabannezhad, Niloofar Jaafari, and Majid Safa
- Subjects
P53 ,Tumor suppressor ,P53 Regulation ,DNA damage response ,Prognosis ,Diagnosis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract The p53 protein, encoded by the TP53 gene, serves as a critical tumor suppressor, playing a vital role in maintaining genomic stability and regulating cellular responses to stress. Dysregulation of p53 is frequently observed in hematological malignancies, significantly impacting disease progression and patient outcomes. This review aims to examine the regulatory mechanisms of p53, the implications of TP53 mutations in various hematological cancers, and emerging therapeutic strategies targeting p53. We conducted a comprehensive literature review to synthesize recent findings related to p53's multifaceted role in hematologic cancers, focusing on its regulatory pathways and therapeutic potential. TP53 mutations in hematological malignancies often lead to treatment resistance and poor prognosis. Current therapeutic strategies, including p53 reactivation and gene therapy, show promise in improving treatment outcomes. Understanding the intricacies of p53 regulation and the consequences of its mutations is essential for developing effective diagnostic and therapeutic strategies in hematological malignancies, ultimately enhancing patient care and survival.
- Published
- 2024
- Full Text
- View/download PDF
44. Expression, DNA methylation pattern and transcription factor EPB41L3 in gastric cancer: a study of 262 cases
- Author
-
Mengdi Cai, Haonan Guo, Dong Wang, Tie Zhao, Xiao Liang, Jiaqi Li, XiaoBo Cui, Songbin Fu, and Jingcui Yu
- Subjects
Stomach neoplasms ,Tumor suppressor ,DNA methylation ,Promoter region ,Transcription factors ,Non-CpGs methylation ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Purpose DNA methylation prominently inactivates tumor suppressor genes and facilitates oncogenesis. Previously, we delineated a chromosome 18 deletion encompassing the erythrocyte membrane protein band 4.1-like 3 (EPB41L3) gene, a progenitor for the tumor suppressor that is differentially expressed in adenocarcinoma of the lung-1 (DAL-1) in gastric cancer (GC). Methods Our current investigation aimed to elucidate EPB41L3 expression and methylation in GC, identify regulatory transcription factors, and identify affected downstream pathways. Immunohistochemistry demonstrated that DAL-1 expression is markedly reduced in GC tissues, with its downregulation serving as an independent prognostic marker. Results High-throughput bisulfite sequencing of 70 GC patient tissue pairs revealed that higher methylation of non-CpGs in the EPB41L3 promoter was correlated with more malignant tumor progression and higher-grade tissue classification. Such hypermethylation was shown to diminish DAL-1 expression, thus contributing to the malignancy of GC phenotypes. The DNA methyltransferase inhibitor 5-aza-2’-deoxycytidine (5-aza-CdR) was found to partially restore DAL-1 expression. Moreover, direct binding of the transcription factor CDC5L to the upstream region of the EPB41L3 promoter was identified via chromosome immunoprecipitation (ChIP)-qPCR and luciferase reporter assays. Immunohistochemistry confirmed the positive correlation between CDC5L and DAL-1 protein levels. Subsequent RNA-seq analysis revealed that DAL-1 significantly influences the extracellular matrix and space-related pathways. GC cell RNA-seq post-5-Aza-CdR treatment and single-cell RNA-seq data of GC tissues confirmed the upregulation of AREG and COL17A1, pivotal tumor suppressors, in response to EPB41L3 demethylation or overexpression in GC epithelial cells. Conclusion In conclusion, this study elucidates the association between non-CpG methylation of EPB41L3 and GC progression and identifies the key transcription factors and downstream molecules involved. These findings enhance our understanding of the role of EPB41L3 in gastric cancer and provide a solid theoretical foundation for future research and potential clinical applications.
- Published
- 2024
- Full Text
- View/download PDF
45. SEC14L3 knockdown inhibited clear cell renal cell carcinoma proliferation, metastasis and sunitinib resistance through an SEC14L3/RPS3/NFκB positive feedback loop
- Author
-
Ziming Jiang, Guangcan Yang, Guangchun Wang, Jiayi Wan, Yifan Zhang, Wei Song, Houliang Zhang, Jinliang Ni, Haipeng Zhang, Ming Luo, Keyi Wang, and Bo Peng
- Subjects
SEC14L3 ,RPS3 ,NFκB ,Nanoparticles ,Clear cell renal cell carcinoma ,Tumor suppressor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Clear cell renal cell carcinoma (ccRCC) arises from the renal parenchymal epithelium and is the predominant malignant entity of renal cancer, exhibiting increasing incidence and mortality rates over time. SEC14-like 3 (SEC14L3) has emerged as a compelling target for cancer intervention; nevertheless, the precise clinical implications and molecular underpinnings of SEC14L3 in ccRCC remain elusive. Methods By leveraging clinical data and data from the TCGA-ccRCC and GEO datasets, we investigated the association between SEC14L3 expression levels and overall survival rates in ccRCC patients. The biological role and mechanism of SEC14L3 in ccRCC were investigated via in vivo and in vitro experiments. Moreover, siRNA-SEC14L3@PDA@MUC12 nanoparticles (SSPM-NPs) were synthesized and assessed for their therapeutic potential against SEC14L3 through in vivo and in vitro assays. Results Our investigation revealed upregulated SEC14L3 expression in ccRCC tissues, and exogenous downregulation of SEC14L3 robustly suppressed the malignant traits of ccRCC cells. Mechanistically, knocking down SEC14L3 facilitated the ubiquitination-mediated degradation of ribosomal protein S3 (RPS3) and augmented IκBα accumulation in ccRCC. This concerted action thwarted the nuclear translocation of P65, thereby abrogating the activation of the nuclear factor kappa B (NFκB) signaling pathway and impeding ccRCC cell proliferation and metastasis. Furthermore, diminished SEC14L3 levels exerted a suppressive effect on NFKB1 expression within the NFκB signaling cascade. NFKB1 functions as a transcriptional regulator capable of binding to the SEC14L3 enhancer and promoter, thereby promoting SEC14L3 expression. Consequently, the inhibition of SEC14L3 expression was further potentiated, thus forming a positive feedback loop. Additionally, we observed that downregulation of SEC14L3 significantly increased the sensitivity of ccRCC cells to sunitinib. The evaluation of SSPM-NPs nanotherapy highlighted its effectiveness in combination with sunitinib for inhibiting ccRCC growth. Conclusion Our findings not only underscore the promise of SEC14L3 as a therapeutic target but also unveil an SEC14L3/RPS3/NFκB positive feedback loop that curtails ccRCC progression. Modulating SEC14L3 expression to engage this positive feedback loop might herald novel avenues for ccRCC treatment.
- Published
- 2024
- Full Text
- View/download PDF
46. Co-expression of P53 and P60-katanin shapes transcriptome dynamics
- Author
-
Şirin Korulu
- Subjects
katna1 ,tumor suppressor ,gene expression profiling ,microtubule severing ,neuronal differentiation ,Science (General) ,Q1-390 - Abstract
Microtubules (MT), essential elements of the cytoskeleton have important roles in the cell such as intracellular cargo transport, cell motility and cell division. They provide support, growth and maintenance of the axonal and dendritic processes in neurons. Microtubule severing proteins such as katanin and spastin have roles in microtubule reconfiguration. Katanin is one of the best characterized severing proteins and is composed of catalytic subunit p60-katanin and regulatory subunit p80-katanin. The microtubule severing mechanism of p60-katanin has been depicted in detail, but how p60-katanin itself is regulated is still little-known. p53 is an important protein between proliferation and differentiation. It regulates different cellular mechanisms such as cell cycle arrest, senescence, differentiation, and apoptosis. p53 controls proliferation in dividing cells and is related to differentiation by means of affecting neuronal process length in non-dividing neurons. Both p53 and p60-katanin have critical roles in proliferation and differentiation separately. Moreover, these proteins were shown to physically interact, but their combined effect remains unclear. To this aim, the current study reveals the effects of p53 – p60-katanin co-expression on transcriptome of the fibroblast cells. Data indicated that the transcriptome of many different pathways such as actin regulation, neuroactive ligand-receptor interaction, and serotonergic synapses pathways were altered under p53 – p60-katanin co-expression conditions. Exploring combined effect of p53 and p60-katanin will help in design of new studies to better understand not only microtubule regulation but also neurodegenerative diseases that are linked to the reactivation of cell cycle and neuronal damage where two of these players take place.
- Published
- 2024
- Full Text
- View/download PDF
47. Epigenetic silencing of ZIC4 unveils a potential tumor suppressor role in pediatric choroid plexus carcinoma
- Author
-
Dina Hesham, Amal Mosaab, Nada Amer, Nouran Al-Shehaby, Sameh Magdeldin, Ahmed Hassan, Hristo Georgiev, Hisham Elshoky, Mona Rady, Khaled Abou Aisha, Ola Sabet, and Shahenda El-Naggar
- Subjects
ZIC4 ,Pediatric choroid plexus carcinoma ,Epigenetic silencing ,Tumor suppressor ,Transcriptomic profiling ,Proteomic profiling ,Medicine ,Science - Abstract
Abstract Zic family member ZIC4 is a transcription factor that has been shown to be silenced in several cancers. However, understanding the regulation and function of ZIC4 in pediatric choroid plexus tumors (CPTs) remained limited. This study employed data mining and bioinformatics analysis to investigate the DNA methylation status of ZIC4 in CPTs and its correlation with patient survival. Our results unveiled ZIC4 methylation as a segregating factor, dividing CPT cohorts into two clusters, with hyper-methylation linked to adverse prognosis. Hyper-methylation of ZIC4 was confirmed in a choroid plexus carcinoma-derived cell line (CCHE-45) by bisulfite sequencing. Furthermore, our study demonstrated that demethylating agent and a histone methyltransferase inhibitor could reverse ZIC4 silencing. RNA sequencing and proteomic analysis showed that ZIC4 over-expression influenced genes and proteins involved in immune response, antigen processing and presentation, endoplasmic reticulum stress, and metabolism. Functionally, re-expressing ZIC4 negatively impacted cell proliferation and migration. Ultimately, these findings underscore ZIC4 hyper-methylation as a prognostic marker in CPTs and shed light on potential mechanisms underlying its tumor suppressor role in CPC. This insight paves the way for novel therapeutic targets in treating aggressive CPTs.
- Published
- 2024
- Full Text
- View/download PDF
48. ARMCX1 inhibits lung adenocarcinoma progression by recruiting FBXW7 for c-Myc degradation
- Author
-
Zhe Hu, Yilin Wu, Xiaoou Sun, Yanli Tong, Houkuang Qiu, and Enqing Zhuo
- Subjects
ARMCX1 ,c-Myc ,FBXW7 ,Lung adenocarcinoma ,Tumor suppressor ,Ubiquitination ,Biology (General) ,QH301-705.5 - Abstract
Abstract Background Armadillo Repeat Containing X-Linked 1 (ARMCX1), a member of the ARM Repeat X-linked protein family, exerts inhibitory function in various tumors. However, its biological role in lung adenocarcinoma (LUAD) and the underlying molecular mechanisms require further exploration. Methods LUAD tissue microarrays and bioinformatic databases were used to evaluate the relationship between ARMCX1 and clinicopathological features. The influence of ARMCX1 on LUAD cell proliferation, migration, and invasion in vitro was determined by colony formation, CCK-8, EdU incorporation, cell cycle, wound healing, and Transwell assays. The impact of ARMCX1 on LUAD cell growth and metastasis in vivo was determined by subcutaneously transplanted tumor and pulmonary metastasis assays. Western blot, immunoprecipitation, immunofluorescence, cycloheximide, and proteasome inhibitor assays were finally conducted to explore the potential underlying molecular mechanisms. Results ARMCX1 expression was downregulated in clinical LUAD samples due to which patient prognoses were poor. Functional experiments indicated that ARMCX1 overexpression inhibited the growth and metastasis of LUAD cells in vitro and in vivo. The molecular mechanism suggested that ARMCX1 recruits the E3 ubiquitin ligase FBXW7 for mediating ubiquitinated degradation of c-Myc, suppressing its nuclear accumulation, and ultimately inactivating cell cycle and epithelial-mesenchymal transition (EMT) signals. Conclusion ARMCX1 inhibits LUAD cell proliferation and metastasis by interacting with c-Myc and enhancing its ubiquitination and degradation. Consequently, it can act as a tumor suppressor in this disease. These results suggest that ARMCX1 is a potential target in the treatment of LUAD.
- Published
- 2024
- Full Text
- View/download PDF
49. The up-regulation of SYNCRIP promotes the proliferation and tumorigenesis via DNMT3A/p16 in colorectal cancer
- Author
-
Chenglong Li, Tailiang Lu, Hongxi Chen, Zhige Yu, and Chaowu Chen
- Subjects
Colorectal cancer ,hnRNPs ,DNA methyltransferases ,Tumor suppressor ,Tumor growth ,Medicine ,Science - Abstract
Abstract Heterogeneous nuclear ribonucleoproteins (hnRNPs), a group of proteins that control gene expression, have been implicated in many post-transcriptional processes. SYNCRIP (also known as hnRNP Q), a subtype of hnRNPs, has been reported to be involved in mRNA splicing and translation. In addition, the deregulation of SYNCRIP was found in colorectal cancer (CRC). However, the role of SYNCRIP in regulating CRC growth remains largely unknown. Here, we found that SYNCRIP was highly expressed in colorectal cancer by analyzing TCGA and GEPIA database. Furthermore, we confirmed the expression of SYNCRIP expression in CRC tumor and CRC cell lines. Functionally, SYNCRIP depletion using shRNA in CRC cell lines (SW480 and HCT 116) resulted in increased caspase3/7 activity and decreased cell proliferation, as well as migration. Meanwhile, overexpression of SYNCRIP showed opposite results. Mechanistically, SYNCRIP regulated the expression of DNA methyltransferases (DNMT) 3A, but not DNMT1 or DNMT3B, which affected the expression of tumor suppressor, p16. More importantly, our in vivo experiments showed that SYNCRIP depletion significantly inhibited colorectal tumor growth. Taken all together, our results suggest SYNCRIP as a potent therapeutic target in colorectal cancer.
- Published
- 2024
- Full Text
- View/download PDF
50. High Expression of ZNF208 Predicts Better Prognosis and Suppresses the Tumorigenesis of Breast Cancer.
- Author
-
Wei, Jing, Jiao, Fangzheng, Wang, Xiaoya, Qiao, Yifan, Yuan, Zihan, Liu, Fang, Fang, Yan, and Pan, Yanfang
- Subjects
- *
BREAST cancer prognosis , *PREDICTIVE tests , *T-test (Statistics) , *RESEARCH funding , *BREAST tumors , *FISHER exact test , *CANCER patients , *DESCRIPTIVE statistics , *GENE expression , *DNA methylation , *GENES , *KAPLAN-Meier estimator , *LOG-rank test , *CARCINOGENESIS , *COMPARATIVE studies , *STAINS & staining (Microscopy) , *DATA analysis software , *SINGLE nucleotide polymorphisms - Abstract
Background: Breast cancer (BRCA), the hormone related malignant tumor, is well-known for poor prognosis. ZNF208 mainly acts as a transcription factor in various tumors, and the single nucleotide polymorphisms (SNPs) of ZNF208 are related to telomere length. Nevertheless, its role in breast tumorigenesis is largely unknown. Methods: We systematically investigated the gene expression, prognostic value, and promoter methylation of ZNF208 in BRCA with Gene Expression Profiling Interactive Analysis (GEPIA) and DNA Methylation Interactive Visualization Database (DNMIVD). Meanwhile, we clarified the association of ZNF208 with tumor-infiltrating immune cells (TICs) from Tumor Immune Estimation Resource (TIMER). Furthermore, we determined the biological process and functional enrichment from Cancer single-cell state atlas (CancerSEA). Finally, we verified our results with prognostic analysis and immunohistochemistry (IHC) assay. Results: We discovered that ZNF208 was downregulated in breast cancer, and low expression of ZNF208 predicted worse prognosis of BRCA patients. The promoter methylation level of ZNF208 was obviously increased, and ZNF208 was associated with TlCs in BRCA. In addition, ZNF208 could inhibit the metastasis and invasion biological processes, and regulate the MAPK and RAS signaling pathways in BRCA. Conclusion: Our findings illustrate that ZNF208 can function as a tumor suppressor and predict prognosis of breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.