1
|
Shu Y, Cai X, Yang X, Yang Y, Ge L. Demethylated miR-184 regulates EPB41L5 and downregulates Notch signaling to inhibit metastasis in colorectal cancer. J Mol Histol 2025; 56:152. [PMID: 40342066 DOI: 10.1007/s10735-025-10434-2] [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: 12/31/2024] [Accepted: 04/20/2025] [Indexed: 05/11/2025]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality worldwide, with metastasis being a major contributor to poor prognosis. MicroRNA-184 (miR-184) has been implicated in the progression of various cancers, but its role in CRC metastasis remains poorly defined. This study investigated the effects of miR-184 promoter demethylation on EPB41L5 expression and Notch signaling in CRC. SW620 human colon carcinoma cells were treated with the DNA methylation inhibitor 5-Aza for 96 h. Methylation status was assessed via bisulfite sequencing, and gene expression was evaluated using qRT-PCR and Western blotting. Functional assays were conducted to assess cell proliferation, apoptosis, migration, and invasion. 5-Aza treatment significantly decreased miR-184 promoter methylation, leading to increased miR-184 expression. This upregulation suppressed CRC cell migration and invasion, induced G2/M cell cycle arrest, and promoted apoptosis. Mechanistically, miR-184 inhibited EPB41L5 expression, thereby downregulating the Notch signaling pathway and modulating epithelial-mesenchymal transition markers. High EPB41L5 expression in CRC tissues was associated with worse prognosis. These findings suggest that demethylated miR-184 inhibits CRC metastasis by targeting the EPB41L5/Notch signaling axis. This regulatory pathway may serve as a novel prognostic biomarker and therapeutic target, with potential clinical implications for the prevention and treatment of metastatic colorectal cancer.
Collapse
Affiliation(s)
- Yin Shu
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Ürümqi, 830011, Xinjiang, China
| | - Xiaohui Cai
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Ürümqi, 830011, Xinjiang, China
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Xinhui Yang
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Ürümqi, 830011, Xinjiang, China
| | - Yuping Yang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Lei Ge
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Ürümqi, 830011, Xinjiang, China.
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| |
Collapse
|
2
|
Zhou Y, Chen X, Zu X. ZBTB7A as a therapeutic target for cancer. Biochem Biophys Res Commun 2024; 736:150888. [PMID: 39490153 DOI: 10.1016/j.bbrc.2024.150888] [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: 08/13/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
ZBTB7A, alternatively referred to Pokemon, FBI-1, LRF, and OCZF, is classified as a member of POK/ZBTB protein family of transcriptional repressors. ZBTB7A binds to targeted DNA via C-terminal zinc fingers and recruits co-compression complexes through N-terminal BTB ⁄ POZ domain to impede transcription. ZBTB7A regulates a range of fundamental biological processes such as cell proliferation, differentiation and apoptosis, B- and T-lymphocyte fate determination and thymic insulin expression and self-tolerance. Accumulating evidence has demonstrated an important role of ZBTB7A in the initiation and advancement of tumors, thus making ZBTB7A emerge as an appealing target. This review examines the functions and regulatory mechanisms of ZBTB7A in a range of common solid tumors, including hepatocellular carcinoma, breast cancer, prostate cancer and lung cancer, as well as hematological malignancies. Notably, the review concludes with a summary of the recent applications of targeting ZBTB7A in clinical treatments through gene silencing, immunotherapy and chemotherapeutic approaches to halt or slow tumor progression. We focus on the functional role and regulatory mechanisms of ZBTB7A in cancer with the goal of providing new insights for the development of more effective cancer therapeutic strategies.
Collapse
Affiliation(s)
- Ying Zhou
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xisha Chen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, China.
| |
Collapse
|
3
|
Zhang Z, Wang L, Zhang F, Jing S, Cen M. Functional mechanism and clinical implications of mir-1271-5p in pilon fracture healing processes. J Orthop Surg Res 2024; 19:782. [PMID: 39578827 PMCID: PMC11583746 DOI: 10.1186/s13018-024-05291-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Pilon fractures are challenging to treat and carry a risk of delayed healing. MicroRNA (miRNA) is closely associated with various diseases due to its ability to regulate gene expression. Consequently, this study aimed to examine the connection between miR-1271-5p expression levels and pilon fracture healing processes, while also exploring the underlying mechanisms. The objective of this research was to provide valuable insights for the future clinical treatment of pilon fractures. MATERIALS Venous blood samples were obtained for RNA extraction from patients with normal healing (n = 107) or delayed healing (n = 45) of pilon fractures. The expression levels of miR-1271-5p were measured using qRT-PCR. MiR-1271-5p and ZBTB7A biological functions in MC3T3-E1 cells were examined using the Cell Counting Kit-8 (CCK-8), flow cytometry, and qRT-PCR. Finally, an investigation into the underlying mechanisms was carried out using a dual luciferase reporter assay. RESULTS This study found that, compared to those who healed normally, patients who experienced delayed healing of pilon fractures had significantly higher expression of miR-1271-5p. This suggests that miR-1271-5p may be an indicator for delayed healing in pilon fractures. Moreover, the upregulation of miR-1271-5p may result in a reduction of ZBTB7A expression, which is thought to mediate the effects of miR-1271-5p on MC3T3-E1 cell activities. CONCLUSIONS MiR-1271-5p was involved in the healing processes of pilon fractures via targeting ZBTB7A. MiR-1271-5p was a possible target for the therapy of pilon fractures.
Collapse
Affiliation(s)
- Zhihan Zhang
- Department of Orthopaedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Li Wang
- Department of Joint and Trauma Surgery, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Fangyuan Zhang
- Department of Trauma Surgery, First Hospital of Qinhuangdao, Qinhuangdao, 066000, China
| | - Shaochun Jing
- Department of Traumatic Arthropathy, The Second Hospital of Qinhuangdao, No. 133, Chaoyang South Street, Changli County, Qinhuangdao City, 066000, Hebei Province, China.
| | - Meini Cen
- Department of Rehabilitation Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2nd Road, Youjiang District, Baise, 533000, Guangxi, China.
| |
Collapse
|
4
|
Tang J, Chen L, Chang Y, Hang D, Chen G, Wang Y, Feng L, Xu M. ZBTB7A interferes with the RPL5-P53 feedback loop and reduces endoplasmic reticulum stress-induced apoptosis of pancreatic cancer cells. Mol Carcinog 2024; 63:1783-1799. [PMID: 38896079 DOI: 10.1002/mc.23772] [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: 12/22/2023] [Revised: 05/25/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024]
Abstract
Endoplasmic reticulum (ER) stress is a primary mechanism leading to cell apoptosis, making it of great research interests in cancer management. This study delves into the function of ribosomal protein L5 (RPL5) in ER stress within pancreatic cancer (PCa) cells and investigates its regulatory mechanisms. Bioinformatics predictions pinpointed RPL5 as an ER stress-related gene exhibiting diminished expression in PCa. Indeed, RPL5 was found to be poorly expressed in PCa tissues and cells, with this reduced expression correlating with an unfavorable prognosis. Moreover, RPL5 overexpression led to heightened levels of p-PERK, p-eIF2α, and CHOP, bolstering the proapoptotic effect of Tunicamycin, an ER stress activator, on PCa cells. Additionally, the RPL5 overexpression curbed cell proliferation, migration, and invasion. Tunicamycin enhanced the binding between RPL5 and murine double minute 2 (MDM2), thus suppressing MDM2-mediated ubiquitination and degradation of P53. Consequently, P53 augmentation intensified ER stress, which further enhanced the binding between RPL5 and MDM2 through PERK-dependent eIF2α phosphorylation, thereby establishing a positive feedback loop. Zinc finger and BTB domain containing 7A (ZBTB7A), conspicuously overexpressed in PCa samples, repressed RPL5 transcription, thereby reducing P53 expression. Silencing of ZBTB7A heightened ER stress and subdued the malignant attributes of PCa cells, effects counteracted upon RPL5 silencing. Analogous outcomes were recapitulated in vivo employing a xenograft tumor mouse model, where ZBTB7A silencing dampened the tumorigenic potential of PCa cells, an effect reversed by additional RPL5 silencing. In conclusion, this study suggests that ZBTB7A represses RPL5 transcription, thus impeding the RPL5-P53 feedback loop and mitigating ER-induced apoptosis in PCa cells.
Collapse
Affiliation(s)
- Jie Tang
- Department of Gastroenterology, Shanghai Hongkou District Jiangwan Hospital, Shanghai, P.R. China
| | - Lingling Chen
- Department of Gastroenterology, Shanghai Pudong New Area People's Hospital, Shanghai, P.R. China
| | - Yunli Chang
- Department of Gastroenterology, Shanghai Pudong New Area People's Hospital, Shanghai, P.R. China
| | - Dongyun Hang
- Department of Gastroenterology, Shanghai Pudong New Area People's Hospital, Shanghai, P.R. China
| | - Guoyu Chen
- Department of Gastroenterology, Shanghai Pudong New Area People's Hospital, Shanghai, P.R. China
| | - Ying Wang
- Department of Gastroenterology, Shanghai Pudong New Area People's Hospital, Shanghai, P.R. China
| | - Lingmei Feng
- Department of Gastroenterology, Shanghai Pudong New Area People's Hospital, Shanghai, P.R. China
| | - Ming Xu
- Department of Gastroenterology, Shanghai Pudong New Area People's Hospital, Shanghai, P.R. China
| |
Collapse
|
5
|
Yu L, Hu X, Zhu H. Predictive value of procollagen c-protease enhancer protein on the prognosis of glioma patients. Heliyon 2024; 10:e28089. [PMID: 38533063 PMCID: PMC10963382 DOI: 10.1016/j.heliyon.2024.e28089] [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: 10/23/2023] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Procollagen c-protease enhancer protein (PCOLCE) performs an essential action in improving the recreation of procollagen c-protease and promoting the reconstruction of extracellular matrix. High PCOLCE expression was associated with a negative prognosis of stomach cancer, ovarian cancer, and osteosarcoma. The goal of this work is to investigate the function of PCOLCE in glioma. Multiple bioinformatics techniques have been employed to investigate the roles of PCOLCE in glioma, consisting of the correlation between PCOLCE and prognosis, immune checkpoints, immune cell infiltrates, and tumor microenvironment (TME). The gene ontology (GO) annotations and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used to assess the potential function of PCOLCE in glioma. PCOLCE was found to be increased in glioma. We revealed that PCOLCE was a potential prognostic factor and related to tumor grade. Up-regulated PCOLCE was related to poor prognosis in lower-grade glioma (LGG), glioblastoma multiforme (GBM), and recurrent glioma. PCOLCE was correlated with immune cell infiltration, particularly B cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells (DCs) in LGG, and DCs infiltration in GBM. PCOLCE was co-expressed with many genes related to the immune and the immune checkpoint. In addition, glioma patients with low expression of PCOLCE had a higher response to the immunological checkpoint blockade (ICB). Additionally, PCOLCE may exert its roles via several immune-related biological processes or pathways, such as leukocyte migration, activation of T cells, adaptive immune response, neutrophil-mediated immunity, NF-κB, and TNF signaling pathways. In conclusion, PCOLCE may be a new immune-related gene and regulate tumor development through immunological pathways.
Collapse
Affiliation(s)
- Luli Yu
- Department of Neurosurgery, Shangrao People's Hospital, Shangrao, 334000, Jiangxi Province, China
| | - Xinyao Hu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hua Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| |
Collapse
|
6
|
Lofiego MF, Piazzini F, Caruso FP, Marzani F, Solmonese L, Bello E, Celesti F, Costa MC, Noviello T, Mortarini R, Anichini A, Ceccarelli M, Coral S, Di Giacomo AM, Maio M, Covre A. Epigenetic remodeling to improve the efficacy of immunotherapy in human glioblastoma: pre-clinical evidence for development of new immunotherapy approaches. J Transl Med 2024; 22:223. [PMID: 38429759 PMCID: PMC10908027 DOI: 10.1186/s12967-024-05040-x] [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: 12/22/2023] [Accepted: 02/24/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a highly aggressive primary brain tumor, that is refractory to standard treatment and to immunotherapy with immune-checkpoint inhibitors (ICI). Noteworthy, melanoma brain metastases (MM-BM), that share the same niche as GBM, frequently respond to current ICI therapies. Epigenetic modifications regulate GBM cellular proliferation, invasion, and prognosis and may negatively regulate the cross-talk between malignant cells and immune cells in the tumor milieu, likely contributing to limit the efficacy of ICI therapy of GBM. Thus, manipulating the tumor epigenome can be considered a therapeutic opportunity in GBM. METHODS Microarray transcriptional and methylation profiles, followed by gene set enrichment and IPA analyses, were performed to study the differences in the constitutive expression profiles of GBM vs MM-BM cells, compared to the extracranial MM cells and to investigate the modulatory effects of the DNA hypomethylating agent (DHA) guadecitabine among the different tumor cells. The prognostic relevance of DHA-modulated genes was tested by Cox analysis in a TCGA GBM patients' cohort. RESULTS The most striking differences between GBM and MM-BM cells were found to be the enrichment of biological processes associated with tumor growth, invasion, and extravasation with the inhibition of MHC class II antigen processing/presentation in GBM cells. Treatment with guadecitabine reduced these biological differences, shaping GBM cells towards a more immunogenic phenotype. Indeed, in GBM cells, promoter hypomethylation by guadecitabine led to the up-regulation of genes mainly associated with activation, proliferation, and migration of T and B cells and with MHC class II antigen processing/presentation. Among DHA-modulated genes in GBM, 7.6% showed a significant prognostic relevance. Moreover, a large set of immune-related upstream-regulators (URs) were commonly modulated by DHA in GBM, MM-BM, and MM cells: DHA-activated URs enriched for biological processes mainly involved in the regulation of cytokines and chemokines production, inflammatory response, and in Type I/II/III IFN-mediated signaling; conversely, DHA-inhibited URs were involved in metabolic and proliferative pathways. CONCLUSIONS Epigenetic remodeling by guadecitabine represents a promising strategy to increase the efficacy of cancer immunotherapy of GBM, supporting the rationale to develop new epigenetic-based immunotherapeutic approaches for the treatment of this still highly deadly disease.
Collapse
Affiliation(s)
| | | | - Francesca Pia Caruso
- BIOGEM Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples "Federico II", Naples, Italy
| | | | - Laura Solmonese
- Center for Immuno-Oncology, University Hospital of Siena, Siena, Italy
| | | | | | - Maria Claudia Costa
- BIOGEM Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples "Federico II", Naples, Italy
| | - Teresa Noviello
- BIOGEM Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Roberta Mortarini
- Human Tumors Immunobiology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Anichini
- Human Tumors Immunobiology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Ceccarelli
- BIOGEM Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Anna Maria Di Giacomo
- University of Siena, Siena, Italy
- Center for Immuno-Oncology, University Hospital of Siena, Siena, Italy
| | - Michele Maio
- University of Siena, Siena, Italy
- Center for Immuno-Oncology, University Hospital of Siena, Siena, Italy
| | | |
Collapse
|