1
|
Shi L, Fu H, Hao Y, Liu C, Zhou F. Zinc finger transcription factor ZNF384 mitigates LPS-induced ferroptosis and inflammation in lung epithelial cells by activating SESN2-mediated autophagy. Toxicol In Vitro 2025; 107:106073. [PMID: 40374019 DOI: 10.1016/j.tiv.2025.106073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Ferroptosis, a type of programmed cell death distinct from apoptosis, is potentially associated with sepsis-triggered acute respiratory distress syndrome (ARDS). ZNF384 is a transcription factor, but its role in ARDS remain unclear. METHODS Blood samples were collected from sepsis-induced ARDS patients and healthy controls. BEAS-2B cells were stimulated with lipopolysaccharide (LPS) to mimic sepsis-induced damaged lung epithelial cell model. Gene and protein expression were elevated utilizing RT-qPCR, western blot, and immunofluorescent staining, respectively. Cell viability and death were evaluated by CCK-8 and flow cytometry. Inflammatory cytokines and oxidative stress markers were measured using ELISA. Intracellular ROS was determined using DCFH-DA staining. Iron concentration was measured using an iron detection kit. Target relationships were confirmed through ChIP and luciferase reporter assays. RESULTS ZNF384 and SESN2 levels were downregulated in sepsis-induced ARDS patients. Overexpression of ZNF384 reduced inflammatory injury, ferroptosis and enhanced autophagy in LPS-stimulated BEAS-2B cells. Mechanistically, ZNF384 served as transcriptional activator of SESN2 to boost autophagy activation. Rescue experiments validated that depletion of SESN2 strikingly reversed the regulatory function of ZNF384 in LPS-induced inflammation, autophagy and ferroptosis. CONCLUSION ZNF384 alleviated LPS-triggered ferroptosis and inflammation in BEAS-2B cells via activating SESN2-mediated autophagy, indicating ZNF384 was a novel target for ARDS treatment.
Collapse
Affiliation(s)
- Lu Shi
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China; Department of Critical Care Medicine, Chongqing Shapingba Traditional Chinese Medicine Hospital, Chongqing 400030, PR China
| | - Hongxue Fu
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Yingting Hao
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Chang Liu
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China.
| | - Fachun Zhou
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
2
|
Zhang B, Matsumoto Y. Integrated Network Analysis Decipher ZNF384-Related miR-20b-5p and miR-424-5p in Colon Adenocarcinoma. Cancer Rep (Hoboken) 2025; 8:e70233. [PMID: 40405535 DOI: 10.1002/cnr2.70233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 04/09/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND ZNF384 is a C2H2-type zinc finger protein (ZNF) which is implicated in DNA double-strand break (DSB) repair through the classical non-homologous end-joining (cNHEJ) pathway. AIMS To clarify the regulatory mechanisms involving ZNF384 in colon adenocarcinoma (COAD). METHODS AND RESULTS First, we conducted a differential expression gene (DEG) analysis of mRNA and lncRNA using TCGA-COAD RNA-Seq data. We also identified ZNF384-related mRNAs through Pearson's correlation coefficient calculation and conducted weighted gene co-expression network analysis (WGCNA) for these genes, leading to the identification of a cluster of 331 genes with strongly positive correlation to tumor, 84 of which overlapped with DEGs. Gene functional analysis showed enrichment of genes in DNA repair, replication fork, and cell cycle checkpoint signaling pathways. Protein-protein interaction (PPI) network analysis of these 84 genes led to the identification of the top 20 key mRNAs. Then we employed three machine learning methods to refine our selection of candidate genes from these intersecting mRNAs. We constructed a competitive endogenous RNA (ceRNA) network and identified two significant intersecting miRNAs, miR-20b-5p and miR-424-5p, which have been shown to act as a tumor suppressor gene and an oncogene, respectively. Additionally, we found that KIF14 and KIF18B are regulated by these two miRNAs in this ceRNA network, particularly in DNA damage repair and cell cycle. Finally, validation using an external dataset from the GEO database confirmed their expression patterns. CONCLUSION The current study clarifies the mechanisms of how miR-20b-5p and miR-424-5p work in colon cancer and underscores their predictive capabilities in colon cancer.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Transdisciplinary Science and Engineering, School of Environment and Society, Institute of Science Tokyo, Tokyo, Japan
- Laboratory for Zero-Carbon Energy, Institute of Integrated Research, Institute of Science Tokyo, Tokyo, Japan
| | - Yoshihisa Matsumoto
- Department of Transdisciplinary Science and Engineering, School of Environment and Society, Institute of Science Tokyo, Tokyo, Japan
- Laboratory for Zero-Carbon Energy, Institute of Integrated Research, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
3
|
Zhang L, Wang J, Gui F, Peng F, Deng W, Zhu Q. METTL3-mediated m6A modification of ZNF384 promotes hepatocellular carcinoma progression by transcriptionally activating ACSM1. Clin Transl Oncol 2025; 27:2256-2268. [PMID: 39342516 DOI: 10.1007/s12094-024-03701-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a lethal disease with a high mortality rate, and its development is influenced by various molecular mechanisms. Zinc finger protein 384 (ZNF384) has been reported to be involved in the progression of several cancers; however, its role in HCC remains elusive. METHODS mRNA expression levels were analyzed by quantitative real-time polymerase chain reaction, while western blotting and immunohistochemistry were performed to validate protein expression. Cell proliferation, apoptosis, and metabolic activities were examined using clonogenicity, flow cytometry, and specific assay kits. A xenograft mouse model was employed to assess the impact of acyl-CoA synthetase medium-chain family member 1 (ACSM1) depletion on HCC cell malignancy in vivo. Chromatin immunoprecipitation assay and dual-luciferase reporter assay were conducted to explore the association between ZNF384 and ACSM1. RESULTS We found that ACSM1 and ZNF384 were significantly upregulated in HCC tissues and cells when compared with normal liver tissues and human liver immortalized cells. Knockdown of ACSM1 inhibited HCC cell proliferation and glucose metabolism and induced cell apoptosis. Furthermore, ACSM1 depletion suppressed the malignant progression of HCC cells in vivo. Our data indicated that ZNF384 transcriptionally activated ACSM1 in HCC cells. Overexpression of ACSM1 reversed the inhibitory effect of ZNF384 depletion on HCC cell malignancy. Further, methyltransferase-like 3 (METTL3) stabilized ZNF384 mRNA through m6A methylation. CONCLUSION METTL3-mediated m6A modification of ZNF384 contributed to the progression of HCC by transcriptionally activating ACSM1. This finding suggests potential therapeutic targets for this devastating disease.
Collapse
Affiliation(s)
- Limei Zhang
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, No.187, Guanlan Street, Shenzhen, 518110, China
| | - Jinfu Wang
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, No.187, Guanlan Street, Shenzhen, 518110, China
| | - Fenfang Gui
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, No.187, Guanlan Street, Shenzhen, 518110, China
| | - Fanzhou Peng
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, No.187, Guanlan Street, Shenzhen, 518110, China
| | - Weiping Deng
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, No.187, Guanlan Street, Shenzhen, 518110, China
| | - Qian Zhu
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, No.187, Guanlan Street, Shenzhen, 518110, China.
| |
Collapse
|
4
|
Kong S, Li J, Pan X, Zhao C, Li Y. ZNF384 and m6A methylation promote the progression of hepatocellular carcinoma by regulating the interaction between LINC00342 and DAPK1. Cell Signal 2025; 129:111666. [PMID: 39961407 DOI: 10.1016/j.cellsig.2025.111666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/05/2024] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with high morbidity and mortality. Many lncRNAs play important regulatory roles in the pathogenesis of HCC, but the mechanism of action of LINC00342 in the progression of HCC remains unclear. In this study, we assessed 24 pairs of HCC tissues and adjacent normal tissues as well as HCC cells and a nude mouse model of HCC. Gene and protein expression was evaluated by flow cytometry, CCK-8, RIP, colony formation assay, and TUNEL staining. This study revealed that LINC00342 was highly expressed in HCC tissues and cells. LINC00342 knockdown significantly inhibited the proliferation and migration of HCC cells, promoted apoptosis, inhibited tumor growth in vivo, and increased the sensitivity of HCC cells to cisplatin. The opposite effect was observed in LINC00342-overexpressing cells. Mechanistically, ZNF384 and m6A methylation can promote the transcription and stability of LINC00342, and LINC00342 can bind to DAPK1, which inhibits Cyt C release and the activation of caspase family proteins to accelerate HCC progression. Our study indicated that the inhibition of LINC00342 expression may represent a new breakthrough for HCC treatment.
Collapse
Affiliation(s)
- Shujia Kong
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Jiaxun Li
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Xin Pan
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Chen Zhao
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China
| | - Yanwen Li
- Intensive Care Unit, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, Yunnan, China.
| |
Collapse
|
5
|
Lainscsek X, Kong W, Rütgen BC, Beck J, Brenig B, Nolte I, Murua Escobar H, Taher L. Transcriptomic profiling in canine B-cell lymphoma supports a synergistic effect of BTK and PI3K inhibitors. Front Vet Sci 2025; 12:1577028. [PMID: 40351764 PMCID: PMC12063356 DOI: 10.3389/fvets.2025.1577028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction B-cell receptor (BCR) signaling has revealed itself as a critical pathway in the pathogenesis of B-cell lymphoma. Within this pathway, the inhibition of Bruton's tyrosine kinase (BTK) or Phosphoinositide 3-kinases (PI3Ks) alone presents encouraging efficacy in the treatment of certain both canine and human hematological malignancies. Methods Here we characterized the effects of the BTK inhibitor Ibrutinib and the PI3K inhibitor AS-605240 as single and combined agents in the canine pre-clinical diffuse large B cell lymphoma (DLBCL) model CLBL-1 by assaying cell proliferation and metabolic activity, and performing RNA-seq to measure gene expression changes. Results We found 2,336 differentially expressed genes (DEGs) across all treatment types and time points relative to the control. The largest number of DEGs were induced by the combination of Ibrutinib and AS-605240. These genes were involved in adaptive immune response, leukotriene D4 metabolic and terms related to regulation of GTP and GTPase mediated signal transduction. Weighted gene co-expression network analysis (WGCNA) detected nine gene modules, five of which were associated with treatment response. Eighteen-percent of genes within these modules were also differentially expressed. Notably, we observed one module that was exclusively associated with the combined treatment whose gene members were related to cellular metabolism, homeostasis signaling, and protein synthesis and regulation. Conclusion Narrowing in on highly connected genes of modules associated with treatment response with large fold changes across treatments which play roles in the main targeted pathways identified PAG1, PRKAR2A, ACACA, FOS, and PRKCA as potential primary candidates of the synergistic treatment effect.
Collapse
Affiliation(s)
- Xenia Lainscsek
- Institute of Biomedical Informatics, Graz University of Technology, Graz, Austria
| | - Weibo Kong
- Clinic for Hematology, Oncology and Palliative Care, Rostock University Medical Center, University of Rostock, Rostock, Germany
| | - Barbara C. Rütgen
- Department for Pathobiology, Clinical Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Julia Beck
- Chronix Biomedical GmbH, Göttingen, Germany
| | - Bertram Brenig
- Institute of Veterinary Medicine, University of Göttingen, Göttingen, Germany
| | - Ingo Nolte
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hugo Murua Escobar
- Clinic for Hematology, Oncology and Palliative Care, Rostock University Medical Center, University of Rostock, Rostock, Germany
- Institute of Medical Genetics, Rostock University Medical Center, University of Rostock, Rostock, Germany
| | - Leila Taher
- Institute of Biomedical Informatics, Graz University of Technology, Graz, Austria
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, University of Rostock, Rostock, Germany
| |
Collapse
|
6
|
Deng Q, Wu L, He J, Wu F, Jiang Z. Identification of autophagy-related immune targets for enhancing immunotherapy in pancreatic cancer aggressiveness. Discov Oncol 2025; 16:382. [PMID: 40126694 PMCID: PMC11933596 DOI: 10.1007/s12672-025-02190-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) presents significant challenges in oncology, with metastasis critically affecting patient outcomes. Autophagy-related genes (ARGs)'s involvement in influencing immune activity and metastasis in PC remains inadequately understood. AIM This study seeks to identify and validate five ARGs that could serve as immune targets, enhancing enhancing Pancreatic cancer metastasis (PCM)'s prognostic models and informing immunotherapy strategies. METHODS ARGs that were diffentially expressed were screened, followed by Cox regression and LASSO analyses to pinpoint five genes linked to overall survival (OS). A prognostic model was developed and validated using ROC curves. Functional analyses, including GO and KEGG, were performed to elucidate ARG mechanisms. Immune infiltration and TFs/microRNA/mRNA networks were assessed to understand ARG-immune cell interactions. Experimental validation employed real-time PCR, IHC, and Western blotting, supported by TCGA data. Functional assays explored RHEB's role in PC, particularly its interaction with LC3. RESULTS Five ARGs (CASP1, RHEB, CHMP2B, MYC, and HDAC6) were identified, contributing to a robust prognostic model where low-risk individuals showed significantly longer OS. The model demonstrated high AUC scores, indicating strong prognostic capability. CD8 T cells and Treg cells' elevated levels were observed in metastatic subjects. RHEB knockdown suppressed cancer cell proliferation and invasion, with a negative correlation between RHEB and LC3, suggesting a role in autophagy-mediated modulation of PC metastasis. CONCLUSION This study introduces a novel prognostic model incorporating five ARGs, highlighting their potential as immune targets for cancer immunotherapy. The negative correlation between RHEB and LC3 suggests a therapeutic pathway for PCM intervention, laying the groundwork for more effective anti-cancer strategies. These findings advance the identification of novel immune targets and signaling pathways, aligning with precision medicine goals in cancer treatment.
Collapse
Affiliation(s)
- Qianxi Deng
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400010, China
- Department of Gastroenterology, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 621000, Sichuan, China
| | - Linju Wu
- Department of Anesthesiology, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, 2621000, Sichuan, China
| | - Jin He
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400010, China
| | - Fan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400010, China
| | - Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
7
|
Yu T, Zhao J, Li Z, Pan C, Liu J, Zheng K, Wang X, Zhang Y. Research Progress on the Role of Zinc Finger Protein in Colorectal Cancer. Cancer Rep (Hoboken) 2025; 8:e70123. [PMID: 40085529 PMCID: PMC11908617 DOI: 10.1002/cnr2.70123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 12/25/2024] [Accepted: 01/07/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Colorectal cancer is one of the most prevalent malignancies worldwide, with a tendency of increasing incidence in developed countries, which poses a significant threat to the patients' physical and mental health. RECENT FINDINGS The process of gene transcription affects the important physiological functions of cells, so the normal expression of transcription factors is an important prerequisite for maintaining cellular homeostasis. Changes in the level of zinc finger proteins, the most prevalent transcription factor, may play an important trigger for the development of colorectal cancer. Different zinc finger proteins play different roles in terms of promoting or inhibiting cancer development. CONCLUSION This paper briefly reviews the classification, functional characteristics, and expression changes of zinc finger proteins in colorectal cancer, it focuses on how they regulate gene transcription, influence on common signaling pathways, and their potential for translational studies and clinical applications. The objective is to stimulate new ideas for their study of colorectal cancer while also providing foundational information to guide drug development and treatment strategies for colorectal cancer patients in clinical settings.
Collapse
Affiliation(s)
- Tang Yu
- The Third Affiliated Hospital of Kunming Medical UniversityKunming Medical UniversityKunmingChina
- Chongqing Nanchuan District People's HospitalChongqing Medical UniversityChongqingChina
| | - Jiumei Zhao
- Chongqing Nanchuan District People's HospitalChongqing Medical UniversityChongqingChina
| | - Ziwei Li
- Department of Gynecology and Obstetrics, Chongqing Health Center for Women and ChildrenWomen and Children's Hospital of Chongqing Medical UniversityChongqingChina
| | - Chenglong Pan
- The Third Affiliated Hospital of Kunming Medical UniversityKunming Medical UniversityKunmingChina
| | - Jialing Liu
- The Third Affiliated Hospital of Kunming Medical UniversityKunming Medical UniversityKunmingChina
| | - Kepu Zheng
- The Third Affiliated Hospital of Kunming Medical UniversityKunming Medical UniversityKunmingChina
| | - Xiaohao Wang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Yan Zhang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| |
Collapse
|
8
|
Wang L, Li N, Chen Y, Qiao Y, Song Y, Zhang X, Zhao H, Ran W, Li G, Xing X. GPSM1 interacts and cooperates with MMP19 to promote proliferation and EMT in colorectal cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119903. [PMID: 39855604 DOI: 10.1016/j.bbamcr.2025.119903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Among patients with colorectal cancer (CRC), metastasis accounts for the majority of deaths, and epithelial-mesenchymal transition (EMT) is important in the metastatic process. However, the mechanism underlying the correlation between the two in CRC is unknown. Here, we verified that a receptor-independent protein, G-protein signaling modulator 1 (GPSM1), was increased in CRC and had a significant positive correlation with matrix metalloproteinase 19 (MMP19). GPSM1 and MMP19 knockdown or overexpression decreased and increased proliferation, migration and invasion of CRC cells, respectively. In addition, overexpression or knockdown of GPSM1 and MMP19 upregulated and inhibited EMT, respectively. Interfering with MMP19 reversed EMT activation via GPSM1 overexpression. Apoptosis was induced by GPSM1 and MMP19 knockdown and activated the caspase3/Bcl-2/Bax signaling pathway. In conclusion, these results support the role of GPSM1 and MMP19 in CRC progression.
Collapse
Affiliation(s)
- Lu Wang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Na Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yang Chen
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yehua Qiao
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaolin Song
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangyan Zhang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Han Zhao
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenwen Ran
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guangqi Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoming Xing
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
9
|
Zhou Y, Wang J, Chen Y, Lin W, Zhou R, Zhao L, Wang H. NRIR promotes immune escape in hepatocellular cancer by regulating IFNγ-induced PD-L1 expression. J Adv Res 2025:S2090-1232(25)00133-X. [PMID: 40023249 DOI: 10.1016/j.jare.2025.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 11/25/2024] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide, with insensitive treatment and poor prognosis; in recent years, breakthroughs in IFN signaling pathway and PDL1/PD1 signaling pathway in HCC immunotherapy research. OBJECTIVES This study aimed to investigate the molecular mechanisms controlling the immune response and immune evasion. METHODS NRIR was identified as a differential gene affecting the IFN signaling pathway and PDL1/PD1 signaling pathway in HCC by bioinformatics, and the function of NRIR was investigated in the HCC cell model and the xenograft mouse model. Quantitative Real-time PCR (qRT-PCR) was used to detect NRIR and PD-L1 mRNAs in hepatocellular carcinoma tissues, and dual luciferase reporter gene assay, fluorescence in situ hybridization, western blot and RNA immunoprecipitation (RIP) to explore the molecular mechanisms between NRIR and target genes. RESULTS In this study, we observed a significant positive correlation between NRIR and PD-L1 expression in HCC, and NRIR upregulated PD-L1 expression in HCC by modulating the IFNγ signaling pathway. We demonstrated that NRIR recruited the transcription factor ZNF384 to initiate CMPK2 transcription. Furthermore, CMPK2 regulates ATP production to modulate STAT1 activation to affect PD-L1 expression. CONCLUSION Our findings revealed the important players of NRIR in regulating PD-L1 expression in HCC and provided new insights for the clinical application of immune-targeted therapies.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Medical Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jing Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuanhang Chen
- Department of Medical Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wandie Lin
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Rui Zhou
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Liang Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Hui Wang
- Department of Medical Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
10
|
Yang Y, He R, Li D, Mu T, Kuang Z, Wang M. The pivotal role of ZNF384: driving the malignant behavior of serous ovarian cancer cells via the LIN28B/UBD axis. Cell Biol Toxicol 2024; 40:100. [PMID: 39562372 PMCID: PMC11576860 DOI: 10.1007/s10565-024-09938-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/16/2024] [Indexed: 11/21/2024]
Abstract
Zinc finger protein 384 (ZNF384) is a highly conserved transcribed gene associated with the development of multiple tumors, however, its role and mechanism in serous ovarian cancer (SOC) are unknown. We first confirmed that ZNF384 was abnormally highly expressed in SOC tissues by bioinformatics analysis and immunohistochemistry. We further used lentivirus packaging and transfection techniques to construct ZNF384 overexpression or knockdown cell lines, and through a series of cell function experiments, gradually verified that ZNF384 promoted a series of malignant behaviors of SOC cell proliferation, migration, and invasion. By establishing a xenotransplantation model in nude mice, it was confirmed that ZNF384 promoted the progress of SOC in vivo. Mechanistically, Overexpression of ZNF384 enhanced the transcriptional activity of Lin-28 homolog B (LIN28B), which promoted the malignant behavior of SOC cells. In addition, LIN28B could regulate the expression of the downstream factor ubiquitin D (UBD) in SOC cells, further promoting the development of SOC. This study shows that ZNF384 aggravates the malignant behavior of SOC cells through the LIN28B/UBD axis, which may be used as a diagnostic biomarker for patients with SOC.
Collapse
Affiliation(s)
- Ye Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Runze He
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Dongxiao Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Tianli Mu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Ziteng Kuang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Min Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
11
|
Chen JP, Diekmann C, Wu H, Chen C, Della Chiara G, Berrino E, Georgiadis KL, Bouwman BAM, Virdi M, Harbers L, Bellomo SE, Marchiò C, Bienko M, Crosetto N. scCircle-seq unveils the diversity and complexity of extrachromosomal circular DNAs in single cells. Nat Commun 2024; 15:1768. [PMID: 38409079 PMCID: PMC10897160 DOI: 10.1038/s41467-024-45972-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 02/08/2024] [Indexed: 02/28/2024] Open
Abstract
Extrachromosomal circular DNAs (eccDNAs) have emerged as important intra-cellular mobile genetic elements that affect gene copy number and exert in trans regulatory roles within the cell nucleus. Here, we describe scCircle-seq, a method for profiling eccDNAs and unraveling their diversity and complexity in single cells. We implement and validate scCircle-seq in normal and cancer cell lines, demonstrating that most eccDNAs vary largely between cells and are stochastically inherited during cell division, although their genomic landscape is cell type-specific and can be used to accurately cluster cells of the same origin. eccDNAs are preferentially produced from chromatin regions enriched in H3K9me3 and H3K27me3 histone marks and are induced during replication stress conditions. Concomitant sequencing of eccDNAs and RNA from the same cell uncovers the absence of correlation between eccDNA copy number and gene expression levels, except for a few oncogenes, including MYC, contained within a large eccDNA in colorectal cancer cells. Lastly, we apply scCircle-seq to one prostate cancer and two breast cancer specimens, revealing cancer-specific eccDNA landscapes and a higher propensity of eccDNAs to form in amplified genomic regions. scCircle-seq is a scalable tool that can be used to dissect the complexity of eccDNAs across different cell and tissue types, and further expands the potential of eccDNAs for cancer diagnostics.
Collapse
Affiliation(s)
- Jinxin Phaedo Chen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden.
- Science for Life Laboratory, Tomtebodavägen 23A, Solna, 17165, Sweden.
| | - Constantin Diekmann
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden
- Science for Life Laboratory, Tomtebodavägen 23A, Solna, 17165, Sweden
| | - Honggui Wu
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, PR China
- School of Life Sciences, Peking University, Beijing, PR China
| | - Chong Chen
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | | | - Enrico Berrino
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, SP142, km 3,95, 10060, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Konstantinos L Georgiadis
- Science for Life Laboratory, Tomtebodavägen 23A, Solna, 17165, Sweden
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Britta A M Bouwman
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden
- Science for Life Laboratory, Tomtebodavägen 23A, Solna, 17165, Sweden
| | - Mohit Virdi
- Human Technopole, Viale Rita Levi-Montalcini 1, 22157, Milan, Italy
| | - Luuk Harbers
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden
- Science for Life Laboratory, Tomtebodavägen 23A, Solna, 17165, Sweden
| | - Sara Erika Bellomo
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, SP142, km 3,95, 10060, Turin, Italy
| | - Caterina Marchiò
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, SP142, km 3,95, 10060, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Magda Bienko
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden.
- Science for Life Laboratory, Tomtebodavägen 23A, Solna, 17165, Sweden.
- Human Technopole, Viale Rita Levi-Montalcini 1, 22157, Milan, Italy.
| | - Nicola Crosetto
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17177, Sweden.
- Science for Life Laboratory, Tomtebodavägen 23A, Solna, 17165, Sweden.
- Human Technopole, Viale Rita Levi-Montalcini 1, 22157, Milan, Italy.
| |
Collapse
|
12
|
Cosgrove BD, Bounds LR, Taylor CK, Su AL, Rizzo AJ, Barrera A, Crawford GE, Hoffman BD, Gersbach CA. Mechanosensitive genomic enhancers potentiate the cellular response to matrix stiffness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.574997. [PMID: 38260455 PMCID: PMC10802421 DOI: 10.1101/2024.01.10.574997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Epigenetic control of cellular transcription and phenotype is influenced by changes in the cellular microenvironment, yet how mechanical cues from these microenvironments precisely influence epigenetic state to regulate transcription remains largely unmapped. Here, we combine genome-wide epigenome profiling, epigenome editing, and phenotypic and single-cell RNA-seq CRISPR screening to identify a new class of genomic enhancers that responds to the mechanical microenvironment. These 'mechanoenhancers' could be active on either soft or stiff extracellular matrix contexts, and regulated transcription to influence critical cell functions including apoptosis, mechanotransduction, proliferation, and migration. Epigenetic editing of mechanoenhancers on rigid materials tuned gene expression to levels observed on softer materials, thereby reprogramming the cellular response to the mechanical microenvironment. These editing approaches may enable the precise alteration of mechanically-driven disease states.
Collapse
Affiliation(s)
- Brian D. Cosgrove
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Lexi R. Bounds
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Carson Key Taylor
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Alan L. Su
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Anthony J. Rizzo
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Alejandro Barrera
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
- Department of Biostatistics and Bioinformatics, Duke University; Durham, NC 27708, USA
| | - Gregory E. Crawford
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
- Department of Pediatrics, Duke University Medical Center; Durham, NC 27708, USA
| | - Brenton D. Hoffman
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Department of Cell Biology, Duke University; Durham, NC 27708, USA
| | - Charles A. Gersbach
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
- Department of Cell Biology, Duke University; Durham, NC 27708, USA
- Department of Surgery, Duke University Medical Center; Durham, NC 27708, USA
| |
Collapse
|
13
|
Yin DP, Zhang H, Teng H, Zhang D, Chen P, Xie L, Liu JS. Overexpressed Gαi1 exerts pro-tumorigenic activity in nasopharyngeal carcinoma. Cell Death Dis 2023; 14:792. [PMID: 38049415 PMCID: PMC10696052 DOI: 10.1038/s41419-023-06308-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/04/2023] [Accepted: 11/14/2023] [Indexed: 12/06/2023]
Abstract
The current study tested the expression and potential functions of Gαi1 in nasopharyngeal carcinoma (NPC). The Cancer Genome Atlas (TCGA) database results demonstrate that Gαi1 transcripts' number in NPC tissues is significantly higher than that in the normal nasal epithelial tissues. Its overexpression correlates with poor survival in certain NPC patients. Moreover, Gαi1 is significantly upregulated in NPC tissues of local primary patients and in different primary human NPC cells. Whereas its expression is relatively low in cancer-surrounding normal tissues and in primary nasal epithelial cells. Genetic silencing (via shRNA strategy) or knockout (via CRISPR-sgRNA method) of Gαi1 substantially suppressed viability, proliferation, cell cycle progression, and migration in primary NPC cells, causing significant caspase-apoptosis activation. Contrarily, ectopic Gαi1 expression exerted pro-tumorigenic activity and strengthened cell proliferation and migration in primary NPC cells. Gαi1 is important for Akt-mTOR activation in NPC cells. Akt-S6K phosphorylation was downregulated after Gαi1 shRNA or KO in primary NPC cells, but strengthened following Gαi1 overexpression. In Gαi1-silenced primary NPC cells, a S473D constitutively-active mutant Akt1 (caAkt1) restored Akt-S6K phosphorylation and ameliorated Gαi1 shRNA-induced proliferation inhibition, migration reduction and apoptosis. Bioinformatics analyses proposed zinc finger protein 384 (ZNF384) as a potential transcription factor of Gαi1. In primary NPC cells, ZNF384 shRNA or knockout (via CRISPR-sgRNA method) decreased Gαi1 mRNA and protein expression, whereas ZNF384 overexpression upregulated it. Importantly, there was an increased binding between ZNF384 protein and the Gαi1 promoter in human NPC tissues and different NPC cells. In vivo studies showed that intratumoral injection of Gαi1-shRNA-expressing adeno-associated virus (AAV) impeded subcutaneous NPC xenograft growth in nude mice. Gαi1 downregulation, Akt-mTOR inactivation, and apoptosis induction were detected in Gαi1-silenced NPC xenograft tissues. Gαi1 KO also effectively inhibited the growth of NPC xenografts in nude mice. Together, overexpressed Gαi1 exerts pro-tumorigenic activity in NPC possibly by promoting Akt-mTOR activation.
Collapse
Affiliation(s)
- De-Pei Yin
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, Children's Hospital of Soochow University, Suzhou, China
| | - Huanle Zhang
- Department of Radiotherapy, Suzhou Ninth People's Hospital, Suzhou, China
| | - Hua Teng
- Department of Otorhinolaryngology Head and Neck Surgery, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing, China
| | - Dan Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Peipei Chen
- Department of Otorhinolaryngology Head and Neck Surgery, Children's Hospital of Soochow University, Suzhou, China.
| | - Lixiao Xie
- Department of Otorhinolaryngology Head and Neck Surgery, Children's Hospital of Soochow University, Suzhou, China.
| | - Ji-Sheng Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
14
|
Gonzalez-Avila G, Sommer B, Flores-Soto E, Aquino-Galvez A. Hypoxic Effects on Matrix Metalloproteinases' Expression in the Tumor Microenvironment and Therapeutic Perspectives. Int J Mol Sci 2023; 24:16887. [PMID: 38069210 PMCID: PMC10707261 DOI: 10.3390/ijms242316887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
The tumor microenvironment (TME) is characterized by an acidic pH and low oxygen concentrations. Hypoxia induces neoplastic cell evasion of the immune surveillance, rapid DNA repair, metabolic reprogramming, and metastasis, mainly as a response to the hypoxic inducible factors (HIFs). Likewise, cancer cells increase matrix metalloproteinases' (MMPs) expression in response to TME conditions, allowing them to migrate from the primary tumor to different tissues. Since HIFs and MMPs are augmented in the hypoxic TME, it is easy to consider that HIFs participate directly in their expression regulation. However, not all MMPs have a hypoxia response element (HRE)-HIF binding site. Moreover, different transcription factors and signaling pathways activated in hypoxia conditions through HIFs or in a HIF-independent manner participate in MMPs' transcription. The present review focuses on MMPs' expression in normal and hypoxic conditions, considering HIFs and a HIF-independent transcription control. In addition, since the hypoxic TME causes resistance to anticancer conventional therapy, treatment approaches using MMPs as a target alone, or in combination with other therapies, are also discussed.
Collapse
Affiliation(s)
- Georgina Gonzalez-Avila
- Laboratorio de Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico;
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico;
| | - Arnoldo Aquino-Galvez
- Laboratorio de Biología Molecular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico;
| |
Collapse
|
15
|
Zhao T, Li X, Li M, Jamil M, Zhang J. Characterization and verification of MMP family members as potential biomarkers in kidney clear cell renal carcinoma. Am J Cancer Res 2023; 13:3941-3962. [PMID: 37818055 PMCID: PMC10560920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 08/13/2023] [Indexed: 10/12/2023] Open
Abstract
Renal cell carcinoma can arise from lesions in the renal epithelium. This particular type of cancer is prevalent in the realm of renal cancers and is associated with an unfavorable prognosis. Among these cases, over 70% are classified as kidney renal clear cell carcinoma (KIRC). Since the underlying causes of KIRC haven't been fully understood, there is an urgent need for deeper investigation into its pathogenesis. Various tools, software, and molecular analysis was used, including Search Tool for the Retrieval of Interacting Genes/Proteins (STRING), Cytoscape, University of ALabama at Birmingham CANcer data analysis Portal (UALCAN), muTarget, Gene Expression Profiling Interactive Analysis (GEPIA), OncoDB, Human Protein Atlas (HPA), cBioPortal, Kaplan-Meier (KM) plotter, Gene Set Enrichment Analysis (GSEA), Tumor IMmune Estimation Resource (TIMER), Encyclopedia of RNA Interactomes (ENCORI), DrugBank, Encyclopedia of RNA Interactomes (RT-qPCR), targeted bisulfide sequencing (bisulfide-seq), and receiver operating curve (ROC) to matrix metallopeptidase (MMP) gene family constituents, with the precise objective of identifying a small set of hub genes. These hub genes hold the potential to be harnessed as molecular biomarkers for KIRC. By performing STRING and CytoHubba analyses of the 24 MMP gene family members, MMP2 (matrix metallopeptidase 2), MMP9 (matrix metallopeptidase 9), MMP14 (matrix metallopeptidase 14), and MMP16 (matrix metallopeptidase 16) were recognized as hub genes having highest degree scores. After conducting an in-depth expression analysis of MMP2, MMP9, MMP14, and MMP16 using various The Cancer Genome Atlas (TCGA) databases and RT-qPCR techniques, these displayed a significant increase in expression at both the mRNA and protein levels within KIRC samples when compared to control samples. The impact of the over expression of MMP2, MMP9, MMP14, and MMP16 also left a distinct mark on the worst overall survival (OS) rates of KIRC patients. Furthermore, a targeted bisulfide-seq investigation unveiled a correlation between promoter hypomethylation patterns and the up-regulation of these key genes in KIRC patients. Additionally, hub genes were involved in various diverse oncogenic pathways. In conclusion, four MMP gene family members, including MMP2, MMP9, MMP14, and MMP16 may serve as therapeutic target and molecular biomarker in KIRC.
Collapse
Affiliation(s)
- Tianyu Zhao
- Central People’s Hospital of ZhanjiangZhanjiang 524000, Guangdong, China
| | - Xue Li
- Central People’s Hospital of ZhanjiangZhanjiang 524000, Guangdong, China
| | - Mingfeng Li
- Central People’s Hospital of ZhanjiangZhanjiang 524000, Guangdong, China
| | - Muhammad Jamil
- PARC Arid Zone Research CenterDera Ismail Khan 29050, Pakistan
| | - Jingyu Zhang
- Central People’s Hospital of ZhanjiangZhanjiang 524000, Guangdong, China
| |
Collapse
|
16
|
Iyer AS, Shaik MR, Raufman JP, Xie G. The Roles of Zinc Finger Proteins in Colorectal Cancer. Int J Mol Sci 2023; 24:10249. [PMID: 37373394 DOI: 10.3390/ijms241210249] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Despite colorectal cancer remaining a leading worldwide cause of cancer-related death, there remains a paucity of effective treatments for advanced disease. The molecular mechanisms underlying the development of colorectal cancer include altered cell signaling and cell cycle regulation that may result from epigenetic modifications of gene expression and function. Acting as important transcriptional regulators of normal biological processes, zinc finger proteins also play key roles in regulating the cellular mechanisms underlying colorectal neoplasia. These actions impact cell differentiation and proliferation, epithelial-mesenchymal transition, apoptosis, homeostasis, senescence, and maintenance of stemness. With the goal of highlighting promising points of therapeutic intervention, we review the oncogenic and tumor suppressor roles of zinc finger proteins with respect to colorectal cancer tumorigenesis and progression.
Collapse
Affiliation(s)
- Aishwarya S Iyer
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mohammed Rifat Shaik
- Department of Medicine, University of Maryland Medical Center Midtown Campus, Baltimore, MD 21201, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- VA Maryland Healthcare System, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Guofeng Xie
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- VA Maryland Healthcare System, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
17
|
Xu F, Yang X, Xue L, Zhang P, Chong T, Sun Y. Clinical significance and biofunction of ZNF384 in renal cell carcinoma discovered by data mining and experimentation. Hum Cell 2023; 36:1214-1217. [PMID: 36882592 DOI: 10.1007/s13577-023-00891-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023]
Affiliation(s)
- Fangshi Xu
- Department of Urology, Shaanxi Provincial People's Hospital, No. 256, Friendship West Road, Xi'an, 710068, Shaanxi, China
| | - XiaoJie Yang
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Five Road, Xi'an, 710004, Shaanxi, China
| | - Li Xue
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Five Road, Xi'an, 710004, Shaanxi, China
| | - Peng Zhang
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Five Road, Xi'an, 710004, Shaanxi, China
| | - Tie Chong
- Department of Urology, Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Five Road, Xi'an, 710004, Shaanxi, China
| | - Yi Sun
- Department of Urology, Shaanxi Provincial People's Hospital, No. 256, Friendship West Road, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
18
|
Meng QX, Wang KN, Li JH, Zhang H, Chen ZH, Zhou XJ, Cao XC, Wang P, Yu Y. ZNF384–ZEB1 feedback loop regulates breast cancer metastasis. Mol Med 2022; 28:111. [PMID: 36100877 PMCID: PMC9469556 DOI: 10.1186/s10020-022-00541-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background Breast cancer has become the most frequently diagnosed cancer worldwide. Increasing evidence indicated that zinc finger proteins (ZNFs), the largest family of transcription factors, contribute to cancer development and progression. Although ZNF384 is overexpressed in several types of human cancer, the role of ZNF384 in breast cancer remains unknown. Therefore, our research focused on ZNF384 regulation of the malignant phenotype of breast cancer and the underlying molecular mechanisms. Methods CCK-8 and colony formation assays were used to evaluate cell proliferation. Transwell and scratch assays were used to evaluate the cell migration and invasion. Chromatin immunoprecipitation (ChIP)-qPCR and luciferase reporter assays were used to confirm the target relationship between ZNF384 and zinc finger E-box binding homeobox 1 (ZEB1). Xenografts were used to monitor the targets in vivo effects. Results We noted that ZNF384 was significantly overexpressed in breast cancer and highlighted the oncogenic mechanism of ZNF384. ZNF384 transactivated ZEB1 expression and induced an epithelial and mesenchymal-like phenotype, resulting in breast cancer metastasis. Furthermore, ZNF384 may be a target of miR-485-5p, and ZEB1 can up-regulate ZNF384 expression by repressing miR-485-5p expression. Together, we unveiled a feedback loop of ZNF384–ZEB1 in breast cancer metastasis. Conclusions The findings suggest that ZNF384 can serve as a prognostic factor and a therapeutic target for breast cancer patients. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00541-1.
Collapse
|