1
|
Xie H, Li J, Lu M, Zhang R, Mao H. miR-1180 Targets FXYD5 to Regulate Pancreatic Cancer Cells Migration and Invasion. Mol Biotechnol 2024; 66:3182-3194. [PMID: 38155284 DOI: 10.1007/s12033-023-00923-8] [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: 08/27/2023] [Accepted: 09/22/2023] [Indexed: 12/30/2023]
Abstract
Pancreatic cancer is a fatal malignancy typically diagnosed in older males and has an aggressive progression. The function of the miR-1180/FXYD5 axis in pancreatic cancer malignant behaviors was investigated. 20 pairs of pancreatic cancer and adjacent normal tissue samples were harvested from pancreatic cancer patients, and qRT-PCR, IHC, and western blot assays were performed, respectively, to detect the mRNA expression and protein levels of miR-1180 or FXYD5. Transwell and scratch assays were conducted to detect the migratory and invasive ability of pancreatic cancer cells; a Dual-luciferase reporter assay was employed to validate miR-1180 targeting FXYD5. miR-1180 targeting FXYD5 to regulate the migratory and invasive ability of pancreatic cancer cells was validated in mouse xenograft tumor models. FXYD5 expression was increased in pancreatic cancer tissue samples than in adjacent normal tissue samples (P < 0.01), and FXYD5 expression exhibited a positive correlation with the migratory and invasive ability of pancreatic cancer cells. miR-1180 targeted FXYD5 and negatively regulated FXYD5. Restoring miR-1180 expression could inhibit the migratory and invasive ability of pancreatic cancer cells (P < 0.01), and this effect could potentially be alleviated by FXYD5 overexpression. The miR-1180/FXYD5 axis positively regulated E-cadherin and negatively regulated MMP2 and MMP9 expression levels. In vivo findings demonstrated that miR-1180 overexpression inhibited tumor growth and lung metastasis (P < 0.05), while FXYD5 overexpression promoted tumor growth and lung metastasis (P < 0.05). In conclusion, the miR-1180 /FXYD5 axis could be involved in pancreatic cancer metastasis through the regulation of EMT and extracellular matrix degradation.
Collapse
Affiliation(s)
- Hongmin Xie
- Department of Gastroenterology, Zhujiang Hospital of Southern Medical University, No. 253 Industrial Road, Guangzhou, 510280, Guangdong, China
- Department of Gastroenterology, Zhongshan Chenxinghai Hospital of Integrated Traditional Chinese and Western Medicine, Zhongshan, 528415, Guangdong, China
| | - Jiaxuan Li
- Department of Gastroenterology, Zhongshan Chenxinghai Hospital of Integrated Traditional Chinese and Western Medicine, Zhongshan, 528415, Guangdong, China
| | - Min Lu
- Department of Gastroenterology, Zhujiang Hospital of Southern Medical University, No. 253 Industrial Road, Guangzhou, 510280, Guangdong, China
| | - Ruijiang Zhang
- Department of Gastroenterology, Zhongshan Chenxinghai Hospital of Integrated Traditional Chinese and Western Medicine, Zhongshan, 528415, Guangdong, China
| | - Hua Mao
- Department of Gastroenterology, Zhujiang Hospital of Southern Medical University, No. 253 Industrial Road, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
2
|
Bai Y, Li L, Li J, Lu X. Association analysis of FXYD5 with prognosis and immunological characteristics across pan-cancer. Heliyon 2024; 10:e30727. [PMID: 38774095 PMCID: PMC11107115 DOI: 10.1016/j.heliyon.2024.e30727] [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: 09/15/2023] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
Background The FXYD domain-containing ion transport regulator 5 (FXYD5) gene is a cancer promoter. However, evidence for an association between FXYD5 and various types of cancer is still lacking. Using multi-omics bioinformatics, our study aimed to reveal the expression distribution, prognostic value, immune infiltration correlation, and molecular functions of FXYD5. Methods Using pan-cancer multi-omics data (including The Cancer Genome Atlas, PrognoScan, Gene Expression Profiling Interactive Analysis, cBioPortal, Gene Expression Omnibus, TIMER and scTIME Portal), we assessed the differences in the expression and prognostic value of FXYD5 in malignant tumors. Furthermore, at the single-cell level, we analyze the expression distribution of FXYD5 across different cell types within the tumor microenvironment, and its relationship with the immune microenvironment. Finally, focusing on ovarian cancer, we conducted preliminary validation of the above findings using cell and molecular biology techniques. Results Our results indicated that FXYD5 was up-regulated in various tumor types and was positively associated with tumor progression. We also revealed that FXYD5 was ubiquitously expressed in microenvironmental cells at the single-cell level, and its upregulation was associated with enhanced immune infiltration, cancer-associated fibroblast infiltration, and dysfunction of tumor-infiltrating cytotoxic T lymphocyte. Additionally, its expression was positively correlated with immune checkpoint genes, DNA mismatch repair genes, MSI (microsatellite instability) and TMB (tumor mutational burden) across various cancers. Its higher expression in cytotoxic T lymphocytes attenuated its ability to predict patient survival with PD-L1 (programmed death-ligand 1) blockade therapy, and FXYD5 was found to be a potential regulator of tumor immune escape and resistance to cancer immunotherapies. Based on GSEA (gene set enrichment analysis) and experimental verification, FXYD5 activated TGF-β/SMAD signaling and drove EMT (epithelial-mesenchymal transition) to promote ovarian cancer progression. Conclusion In summary, our study revealed that FXYD5-TGFβ axis may coregulate the interaction between tumors, CAFs (carcinoma-associated fibroblasts) and immune cells to reshape the tumor immune microenvironment and promote tumorigenesis and tumor progression. Thus, FXYD5 could be used as an immune-related biomarker for diagnosing and predicting the prognosis of multiple cancer types. Therefore, our findings suggest that targeting FXYD5 in TME (tumor microenvironment) may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yang Bai
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Liangdong Li
- Department of Neurosurgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun Li
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Xin Lu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| |
Collapse
|
3
|
Zhao E, Gao K, Xiong J, Liu Z, Chen Y, Yi L. The roles of FXYD family members in ovarian cancer: an integrated analysis by mining TCGA and GEO databases and functional validations. J Cancer Res Clin Oncol 2023; 149:17269-17284. [PMID: 37814066 DOI: 10.1007/s00432-023-05445-z] [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: 08/04/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND The FXYD family of ion transport regulators have emerged as important modulators of cancer progression and metastasis. However, their expression and roles in ovarian cancer (OCa) have not been systematically investigated. METHODS The expression of FXYD genes in OCa was analyzed using data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), as well as independent clinical samples. The prognostic values of FXYD genes were evaluated by Kaplan-Meier and Cox regression analysis. To explore potential mechanisms, bioinformatics approaches including Gene Ontology, KEGG pathway analysis, GSEA and drug sensitivity correlation analysis were performed. OCa cell lines overexpressing FXYD1, FXYD5 or FXYD7 were also generated and their impacts on proliferation, migration and invasion were assessed. RESULTS FXYD1 and FXYD6 were significantly downregulated while FXYD3, FXYD4 and FXYD5 were upregulated in OCa tissues compared to normal tissues. FXYD1, FXYD5 and FXYD7 were independent adverse prognostic factors for OCa patients. Pathway and drug correlation analysis revealed that FXYD1, FXYD5 and FXYD7 genes regulated diverse oncogenic signaling cascades and modulated the response to various chemotherapeutic agents. Overexpression of FXYD1, FXYD5 or FXYD7 enhanced OCa cell motility and invasiveness in vitro. CONCLUSION Our results demonstrate aberrant expression patterns, prognostic values, and oncogenic activities of FXYD genes in OCa. FXYD1, FXYD5 and FXYD7 may serve as biomarkers and therapeutic targets for this disease. Targeting FXYD-mediated signaling represents a promising therapeutic strategy against OCa.
Collapse
Affiliation(s)
- Eryong Zhao
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, 9 Jinsui Road, Guangzhou, 510623, People's Republic of China
| | - Kefei Gao
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, 9 Jinsui Road, Guangzhou, 510623, People's Republic of China
| | - Jian Xiong
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, 9 Jinsui Road, Guangzhou, 510623, People's Republic of China
| | - Zhihong Liu
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, 9 Jinsui Road, Guangzhou, 510623, People's Republic of China
| | - Yuelin Chen
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, 9 Jinsui Road, Guangzhou, 510623, People's Republic of China
| | - Lisha Yi
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, 9 Jinsui Road, Guangzhou, 510623, People's Republic of China.
| |
Collapse
|
4
|
The Role of NF-κB in Endometrial Diseases in Humans and Animals: A Review. Int J Mol Sci 2023; 24:ijms24032901. [PMID: 36769226 PMCID: PMC9917883 DOI: 10.3390/ijms24032901] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The expression of genes of various proinflammatory chemokines and cytokines is controlled, among others, by the signaling pathway of the nuclear factor kappaB (NF-κB) superfamily of proteins, providing an impact on immune system functioning. The present review addresses the influence and role of the NF-κB pathway in the development and progression of most vital endometrial diseases in human and animal species. Immune modulation by NF-κB in endometritis, endometrosis, endometriosis, and carcinoma results in changes in cell migration, proliferation, and inflammation intensity in both the stroma and epithelium. In endometrial cells, the NF-κB signaling pathway may be activated by multiple stimuli, such as bacterial parts, cytokines, or hormones binding to specific receptors. The dysregulation of the immune system in response to NF-κB involves aberrant production of chemokines and cytokines, which plays a role in endometritis, endometriosis, endometrosis, and endometrial carcinoma. However, estrogen and progesterone influence on the reproductive tract always plays a major role in its regulation. Thus, sex hormones cannot be overlooked in endometrial disease physiopathology. While immune system dysregulation seems to be NF-κB-dependent, the hormone-independent and hormone-dependent regulation of NF-κB signaling in the endometrium should be considered in future studies. Future goals in this research should be a step up into clinical trials with compounds affecting NF-κB as treatment for endometrial diseases.
Collapse
|
5
|
Hotta T, Nariai Y, Kajitani N, Kadota K, Maruyama R, Tajima Y, Isobe T, Kamino H, Urano T. Generation of the novel anti-FXYD5 monoclonal antibody and its application to the diagnosis of pancreatic and lung cancer. Biochimie 2023; 208:160-169. [PMID: 36621663 DOI: 10.1016/j.biochi.2023.01.002] [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: 07/03/2022] [Revised: 11/28/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Despite recent advances in cancer treatments, pancreatic cancer has a dismal prognosis globally. Early detection of cancer cells and effective treatments for recalcitrant tumors are required, but the innovative therapeutic tools remain in development. Cancer-specific antigens expressed only on cancer cells may help resolve these problems, and antibodies to such antigens have potential in basic research and clinical applications. To generate specific antibodies that bind to proteins expressed on the surface of pancreatic cancer cells, we immunized mice with human pancreatic cancer MIA PaCa-2 cells, and isolated a hybridoma that produces a monoclonal antibody (mAb), named 12-13.8. This antibody was applied to molecular biological experiments such as immunocytochemistry, immunoblotting, flow cytometry, and immunoprecipitation. In addition, we showed that mAb 12-13.8 could accumulate in tumors, through in vivo experiments using cancer-bearing mice. Immunohistochemical staining of pancreatic and lung tumor tissues indicated that the increase of the staining strength by mAb 12-13.8 positively and inversely correlated with the patients' cancer recurrence and survival rate, respectively. We identified the FXYD5 protein as the target protein of mAb 12-13.8, by a human protein array screening system. The FXYD5 protein is overexpressed in various types of cancer and is modified by O-linked glycosylation. We confirmed the binding of the FXYD5 protein to mAb 12-13.8 by using FXYD5-knockout MIA PaCa-2 cells, and detailed epitope mapping identified amino acid residues 45-52 as the minimal peptide sequence. Our results indicate that mAb 12-13.8 could be a valuable tool for FXYD5 studies, and useful in diagnostic and drug delivery applications for cancer patients.
Collapse
Affiliation(s)
- Takamasa Hotta
- Department of Biochemistry, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan; Division of Medical Oncology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan
| | - Yuko Nariai
- Department of Biochemistry, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan
| | - Naoyo Kajitani
- Department of Experimental Animals, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Shimane, 693-8501, Japan
| | - Kyuichi Kadota
- Department of Pathology, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan
| | - Riruke Maruyama
- Department of Pathology, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan
| | - Yoshitsugu Tajima
- Department of Digestive and General Surgery, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan
| | - Takeshi Isobe
- Division of Medical Oncology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan
| | - Hiroki Kamino
- Department of Biochemistry, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan; mAbProtein Co. Ltd., Izumo, Shimane, 693-8501, Japan.
| | - Takeshi Urano
- Department of Biochemistry, Faculty of Medicine, Shimane University, Izumo, Shimane, 693-8501, Japan; mAbProtein Co. Ltd., Izumo, Shimane, 693-8501, Japan; Vaccines and Therapeutic Antibodies for Emerging Infectious Diseases, Shimane University School of Medicine, Izumo, 693-8501, Japan
| |
Collapse
|
6
|
Niinivirta A, Salo T, Åström P, Juurikka K, Risteli M. Prognostic value of dysadherin in cancer: A systematic review and meta-analysis. Front Oncol 2022; 12:945992. [PMID: 36119538 PMCID: PMC9479204 DOI: 10.3389/fonc.2022.945992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
Cancer is a leading cause of death worldwide and novel prognostic factors are reported with increasing numbers. Systematic reviews and meta-analyses on cumulative research data are crucial in estimating the true prognostic value of proposed factors. Dysadherin (FXYD Domain Containing Ion Transport Regulator 5; FXYD5) is a cell membrane glycoprotein that modulates Na+, K+-ATPase activity and cell-cell adhesion. It is abundantly expressed in a variety of cancer cells, but only in a limited number of normal cells and its levels are increased in many different tumor types. The expression or level of dysadherin has been suggested as an independent predictor for metastasis and poor prognosis by number of studies, yet we lack a definitive answer. In this study, we systematically evaluated the prognostic value of dysadherin in cancer and summarized the current knowledge on the subject. PubMed, Scopus, Web of Science and relevant clinical trial and preprint databases were searched for relevant publications and PRISMA and REMARK guidelines were applied in the process. After a careful review, a total of 23 original research articles were included. In each study, dysadherin was pointed as a marker for poor prognosis. Meta-analyses revealed 3- and 1.5-fold increases in the risk of death (fixed effects HR 3.08, 95% CI 1.88-5.06, RR 1.47, 95% CI 1.06-2.05 on overall survival, respectively) for patients with high (>50%) tumoral FXYD5 level. In many studies, a connection between dysadherin expression or level and metastatic behavior of the cancer as well as inverse correlation with E-cadherin level were reported. Thus, we conclude that dysadherin might be a useful prognostic biomarker in the assessment of disease survival of patients with solid tumors.
Collapse
Affiliation(s)
- Aino Niinivirta
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Oral and Maxillofacial Diseases, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland
- Department of Pathology (HUSLAB), Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland
| | - Pirjo Åström
- Research Unit of Biomedicine, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Krista Juurikka
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Maija Risteli
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| |
Collapse
|
7
|
Tan XP, Xiong BH, Zhang YX, Wang SL, Zuo Q, Li J. FXYD5 promotes sorafenib resistance through the Akt/mTOR signaling pathway in hepatocellular carcinoma. Eur J Pharmacol 2022; 931:175186. [PMID: 35977595 DOI: 10.1016/j.ejphar.2022.175186] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 11/03/2022]
Abstract
Tumor chemoresistance is often a major cause for the failure of chemotherapy. The resistance of hepatocellular carcinoma (HCC) cells to sorafenib significantly limits its therapeutic effect in HCC patients. For the first time, we found that FXYD domain-containing ion transport regulator 5 (FXYD5) is highly expressed in sorafenib-resistant HCC cells. In addition, the protein expression level of FXYD5 was markedly higher in HCC tissues than in paracancerous tissues. Remarkably, downregulation of FXYD5 expression in Huh7/sora cells reversed their resistance to sorafenib. Moreover, overexpression of FXYD5 reduced the sensitivity of HCC cells to sorafenib, while the downregulation of its expression in HCC cells had the opposite effect. We also found abnormal activation of the Akt/mTOR signaling pathway in Huh7/sora cells. Furthermore, MK2206, an Akt inhibitor, was found to significantly increase the sensitivity of HCC cells to sorafenib. More importantly, the expression level of p-Akt was positively correlated with the expression of FXYD5 in HCC tissues. Therefore, mechanistically, FXYD5 enhances the resistance of HCC cells to sorafenib by activating the Akt/mTOR signaling pathway. In conclusion, this study showed that the activation of the FXYD5/Akt/mTOR signaling axis plays key role in the resistance of HCC cells to sorafenib, and FXYD5 may represent a new potential target for HCC therapy.
Collapse
Affiliation(s)
- Xiang-Peng Tan
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Ben-Han Xiong
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yuan-Xu Zhang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Shen-Li Wang
- The Biomedical Translational Research Institute, Jinan University, Guangzhou, China
| | - Qian Zuo
- Department of Breast Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Jing Li
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
8
|
Role of Peritoneal Mesothelial Cells in the Progression of Peritoneal Metastases. Cancers (Basel) 2022; 14:cancers14122856. [PMID: 35740521 PMCID: PMC9221366 DOI: 10.3390/cancers14122856] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022] Open
Abstract
Peritoneal metastatic cancer comprises a heterogeneous group of primary tumors that originate in the peritoneal cavity or metastasize into the peritoneal cavity from a different origin. Metastasis is a characteristic of end-stage disease, often indicative of a poor prognosis with limited treatment options. Peritoneal mesothelial cells (PMCs) are a thin layer of cells present on the surface of the peritoneum. They display differentiated characteristics in embryonic development and adults, representing the first cell layer encountering peritoneal tumors to affect their progression. PMCs have been traditionally considered a barrier to the intraperitoneal implantation and metastasis of tumors; however, recent studies indicate that PMCs can either inhibit or actively promote tumor progression through distinct mechanisms. This article presents a review of the role of PMCs in the progression of peritoneum implanted tumors, offering new ideas for therapeutic targets and related research.
Collapse
|
9
|
Banz-Jansen C, Helweg LP, Kaltschmidt B. Endometrial Cancer Stem Cells: Where Do We Stand and Where Should We Go? Int J Mol Sci 2022; 23:ijms23063412. [PMID: 35328833 PMCID: PMC8955970 DOI: 10.3390/ijms23063412] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/08/2022] [Accepted: 03/19/2022] [Indexed: 02/04/2023] Open
Abstract
Endometrial cancer is one of the most common malignant diseases in women worldwide, with an incidence of 5.9%. Thus, it is the most frequent cancer of the female genital tract, with more than 34,000 women dying, in Europe and North America alone. Endometrial Cancer Stem Cells (CSC) might be drivers of carcinogenesis as well as metastatic and recurrent disease. Therefore, targeting CSCs is of high interest to improve prognosis of patients suffering of advanced or recurrent endometrial cancer. This review describes the current evidence of molecular mechanisms in endometrial CSCs with special emphasis on MYC and NF-κB signaling as well as mitochondrial metabolism. Furthermore, the current status of immunotherapy targeting PD-1 and PD-L1 in endometrial cancer cells and CSCs is elucidated. The outlined findings encourage novel therapies that target signaling pathways in endometrial CSCs as well as immunotherapy as a promising therapeutic approach in the treatment of endometrial cancer to impede cancer progression and prevent recurrence.
Collapse
Affiliation(s)
- Constanze Banz-Jansen
- Department of Gynecology and Obstetrics, and Perinatal Center, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, Burgsteig 13, 33617 Bielefeld, Germany;
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany;
| | - Laureen P. Helweg
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany;
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Correspondence:
| | - Barbara Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld, OWL (FBMB e.V.), Maraweg 21, 33617 Bielefeld, Germany;
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| |
Collapse
|
10
|
Zakrzewski PK. Canonical TGFβ Signaling and Its Contribution to Endometrial Cancer Development and Progression-Underestimated Target of Anticancer Strategies. J Clin Med 2021; 10:3900. [PMID: 34501347 PMCID: PMC8432036 DOI: 10.3390/jcm10173900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023] Open
Abstract
Endometrial cancer is one of the leading gynecological cancers diagnosed among women in their menopausal and postmenopausal age. Despite the progress in molecular biology and medicine, no efficient and powerful diagnostic and prognostic marker is dedicated to endometrial carcinogenesis. The canonical TGFβ pathway is a pleiotropic signaling cascade orchestrating a variety of cellular and molecular processes, whose alterations are responsible for carcinogenesis that originates from different tissue types. This review covers the current knowledge concerning the canonical TGFβ pathway (Smad-dependent) induced by prototypical TGFβ isoforms and the involvement of pathway alterations in the development and progression of endometrial neoplastic lesions. Since Smad-dependent signalization governs opposed cellular processes, such as growth arrest, apoptosis, tumor cells growth and differentiation, as well as angiogenesis and metastasis, TGFβ cascade may act both as a tumor suppressor or tumor promoter. However, the final effect of TGFβ signaling on endometrial cancer cells depends on the cancer disease stage. The multifunctional role of the TGFβ pathway indicates the possible utilization of alterations in the TGFβ cascade as a potential target of novel anticancer strategies.
Collapse
Affiliation(s)
- Piotr K Zakrzewski
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
11
|
Jin M, Zhang H, Yang J, Zheng Z, Liu K. Expression mode and prognostic value of FXYD family members in colon cancer. Aging (Albany NY) 2021; 13:18404-18422. [PMID: 34270462 PMCID: PMC8351680 DOI: 10.18632/aging.203290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022]
Abstract
The FXYD gene family comprises seven members that encode a class of small-membrane proteins characterized by an FXYD motif and interact with Na+/K+-ATPase. Until now, the expression patterns and prognostic roles of the FXYD family in colon cancer (CC) have not been systematically reported. Gene expression, methylation, clinicopathological features and the prognoses of CC patients were obtained from The Cancer Genome Atlas (TCGA) database. The expression feature and prognostic values of FXYD members were identified. Gene set enrichment analysis (GSEA) was performed to explore the potential mechanism underlying the function of the FXYD family in CC. Tumor Immune Estimation Resource (TIMER) and CIBERSORT analysis were used to assess the correlations between FXYD family members and tumor immune infiltrating cells (TIICs). FXYD family members were differentially expressed in CC except for FXYD2. FXYD2, FXYD3 and FXYD4 were revealed as independent prognostic factors for recurrence, while FXYD3 and FXYD7 were identified as prognostic factors for survival according to univariate and multivariate analyses with Cox regression. GSEA revealed that FXYD family members were involved in complicated biological functions underlying cancer progression. TIMER and CIBERSORT analyses showed significant associations between FXYD family genes and TIICs. The present study comprehensively revealed the expression mode and prognostic value of FXYD members in CC, providing insights for further study of the FXYD family as potential clinical biomarkers in CC.
Collapse
Affiliation(s)
- Ming Jin
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Hui Zhang
- Department of Radiation Oncology, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Jun Yang
- Ningbo Diagnostic Pathology Center, Ningbo, China
| | - Zhen Zheng
- Department of Radiation Oncology, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Kaitai Liu
- Department of Radiation Oncology, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| |
Collapse
|
12
|
Juurikka K, Dufour A, Pehkonen K, Mainoli B, Campioni Rodrigues P, Solis N, Klein T, Nyberg P, Overall CM, Salo T, Åström P. MMP8 increases tongue carcinoma cell-cell adhesion and diminishes migration via cleavage of anti-adhesive FXYD5. Oncogenesis 2021; 10:44. [PMID: 34059618 PMCID: PMC8167110 DOI: 10.1038/s41389-021-00334-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/03/2021] [Accepted: 05/14/2021] [Indexed: 12/28/2022] Open
Abstract
Matrix metalloproteinases (MMPs) modify bioactive factors via selective processing or degradation resulting in tumour-promoting or tumour-suppressive effects, such as those by MMP8 in various cancers. We mapped the substrates of MMP8 to elucidate its previously shown tumour-protective role in oral tongue squamous cell carcinoma (OTSCC). MMP8 overexpressing (+) HSC-3 cells, previously demonstrated to have reduced migration and invasion, showed enhanced cell-cell adhesion. By analysing the secretomes of MMP8 + and control cells with terminal amine isotopic labelling of substrates (TAILS) coupled with liquid chromatography and tandem mass spectrometry (LC-MS/MS), we identified 36 potential substrates of MMP8, including FXYD domain-containing ion transport regulator 5 (FXYD5). An anti-adhesive glycoprotein FXYD5 has been previously shown to predict poor survival in OTSCC. Cleavage of FXYD5 by MMP8 was confirmed using recombinant proteins. Furthermore, we detected a loss of FXYD5 levels on cell membrane of MMP8 + cells, which was rescued by inhibition of the proteolytic activity of MMP8. Silencing (si) FXYD5 increased the cell-cell adhesion of control but not that of MMP8 + cells. siFXYD5 diminished the viability and motility of HSC-3 cells independent of MMP8 and similar effects were seen in another tongue cancer cell line, SCC-25. FXYD5 is a novel substrate of MMP8 and reducing FXYD5 levels either with siRNA or cleavage by MMP8 increases cell adhesion leading to reduced motility. FXYD5 being a known prognostic factor in OTSCC, our findings strengthen its potential as a therapeutic target.
Collapse
Affiliation(s)
- K Juurikka
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - A Dufour
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada.,Department of Oral Biological and Medical Sciences, Faculty of Dentistry, Centre for Blood Research, and Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - K Pehkonen
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - B Mainoli
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada
| | - P Campioni Rodrigues
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - N Solis
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, Centre for Blood Research, and Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - T Klein
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, Centre for Blood Research, and Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - P Nyberg
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Biobank Borealis of Northern Finland, Oulu University Hospital, Oulu, Finland
| | - C M Overall
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, Centre for Blood Research, and Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - T Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Department of Oral and Maxillofacial Diseases, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Helsinki University Hospital, Helsinki, Finland.,Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland
| | - P Åström
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland. .,Department of Oral Biological and Medical Sciences, Faculty of Dentistry, Centre for Blood Research, and Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada. .,Research Unit of Biomedicine, Faculty of Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
13
|
Tian M, Wang T, Wang P. Development and Clinical Validation of a Seven-Gene Prognostic Signature Based on Multiple Machine Learning Algorithms in Kidney Cancer. Cell Transplant 2021; 30:963689720969176. [PMID: 33626918 PMCID: PMC7917425 DOI: 10.1177/0963689720969176] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
About a third of patients with kidney cancer experience recurrence or cancer-related progression. Clinically, kidney cancer prognoses may be quite different, even in patients with kidney cancer at the same clinical stage. Therefore, there is an urgent need to screen for kidney cancer prognosis biomarkers. Differentially expressed genes (DEGs) were identified using kidney cancer RNA sequencing data from the Gene Expression Omnibus (GEO) database. Biomarkers were screened using random forest (RF) and support vector machine (SVM) models, and a multigene signature was constructed using the least absolute shrinkage and selection operator (LASSO) regression analysis. Univariate and multivariate Cox regression analyses were performed to explore the relationships between clinical features and prognosis. Finally, the reliability and clinical applicability of the model were validated, and relationships with biological pathways were identified. Western blots were also performed to evaluate gene expression. A total of 50 DEGs were obtained by intersecting the RF and SVM models. A seven-gene signature (RNASET2, EZH2, FXYD5, KIF18A, NAT8, CDCA7, and WNT7B) was constructed by LASSO regression. Univariate and multivariate Cox regression analyses showed that the seven-gene signature was an independent prognostic factor for kidney cancer. Finally, a predictive nomogram was established in The Cancer Genome Atlas (TCGA) cohort and validated internally. In tumor tissue, RNASET2 and FXYD5 were highly expressed and NAT8 was lowly expressed at the protein and transcription levels. This model could complement the clinicopathological characteristics of kidney cancer and promote the personalized management of patients with kidney cancer.
Collapse
Affiliation(s)
- Mi Tian
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tao Wang
- Department of Pathology, Shenyang KingMed Center for Clinical Laboratory Co, Ltd, Shenyang, China
| | - Peng Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
14
|
Liu YK, Jia YJ, Liu SH, Shi HJ, Ma J. Low expression of FXYD5 reverses the cisplatin resistance of epithelial ovarian cancer cells. Histol Histopathol 2021; 36:535-545. [PMID: 33570156 DOI: 10.14670/hh-18-310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the effect of the downregulation of FXYD domain-containing ion transport regulator 5 (FXYD5) on the cisplatin resistance (CisR) of epithelial ovarian cancer (EOC) cells. METHODS A2780-CisR and SKOV3-CisR cells were obtained through repeated administrations of different cisplatin concentrations, and the half-maximal inhibition concentration (IC50) was calculated by MTT assays. After transfection with FXYD5 siRNA-1 and FXYD5 siRNA-2, the IC50 values of the A2780-CisR and SKOV3-CisR cells were also detected by the MTT method. Cell proliferation, migration, invasion and apoptosis were evaluated through 5-ethynyl-2'-deoxyuridine (EdU) DNA synthesis, wound healing, Transwell invasion and Annexin-V-FITC/PI dual-staining assays, respectively. qRT-PCR and Western blotting were conducted to detect mRNA and protein expression. RESULTS Compared with the sensitive parental cells, the A2780-CisR and SKOV3-CisR cells had increased IC50 and FXYD5 expression. FXYD5 siRNA reduced the IC50 value of cisplatin in the A2780-CisR and SKOV3-CisR cells and decreased the expression of ABCG2 (BCRP) and ABCB1 (MDR1). In addition, FXYD5 inhibition reduced the invasion and migration of the A2780-CisR and SKOV3-CisR cells, with upregulation of E-cadherin and downregulation of Snail and Vimentin. Both FXYD5 siRNA-1 and FXYD5 siRNA-2 inhibited the proliferation and promoted the apoptosis of the A2780-CisR and SKOV3-CisR cells with reduced Ki-67 and increased caspase-3. CONCLUSION FXYD5 downregulation may reduce the invasion, migration and EMT formation of EOC cells to increase their sensitivity to cisplatin chemotherapy by inhibiting cell proliferation and promoting cell apoptosis.
Collapse
Affiliation(s)
- Ya-Kun Liu
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Ya-Jing Jia
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shi-Hao Liu
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hong-Jie Shi
- Department of Gynecology, People's Hospital of Tang County, Baoding, Hebei, China
| | - Jing Ma
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
15
|
Liu T, Meng J, Zhang Y. miR‑592 acts as an oncogene and promotes medullary thyroid cancer tumorigenesis by targeting cyclin‑dependent kinase 8. Mol Med Rep 2020; 22:3316-3326. [PMID: 32945439 PMCID: PMC7453674 DOI: 10.3892/mmr.2020.11392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/25/2020] [Indexed: 02/07/2023] Open
Abstract
Medullary thyroid carcinoma (MTC) is a relatively rare subtype of thyroid cancer, accounting for 5‑10% of all cases of thyroid cancer worldwide. Due to the current lack of knowledge regarding the tumorigenesis of MTC, the clinical treatment of MTC remains a challenge. It has been reported that microRNAs (miRNAs) regulate the progression of MTC; however, the regulatory network of miRNAs and the exact underlying mechanisms are not completely understood. In the present study, an miRNA expression profile (GSE40807), consisting of 80 samples, was downloaded and analyzed using Gene Expression Omnibus‑2R to identify differentially expressed miRNAs between MTC and normal samples. miR‑592 expression levels were significantly increased in MTC tissues and cell lines compared with normal tissues and cell lines. Patients with high miR‑592 expression levels exhibited a less favorable prognosis compared with patients with low miR‑592 expression. The results suggested that miR‑592 overexpression promoted TT and MZ‑CRC‑1 cell proliferation in vitro. In addition, miR‑592 negatively regulated cyclin‑dependent kinase 8 (CDK8) via targeted binding in MTC cells. Moreover, co‑transfection of CDK8 overexpression plasmid and miR‑592 mimic reversed miR‑592‑mediated MTC cell proliferation. In conclusion, miR‑592 may serve as an oncogene in MTC by decreasing the expression of CDK8, indicating that the miR‑592/CDK8 axis might serve as a promising therapeutic target for MTC.
Collapse
Affiliation(s)
- Ting Liu
- Department of Nuclear Medicine, The Affiliated Wuhan Central Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jingjing Meng
- Department of Thyroid and Breast Surgery, The Affiliated Wuhan Central Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yu Zhang
- Department of Surgery II, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| |
Collapse
|
16
|
Integrated Transcriptomic Analysis Reveals the Molecular Mechanism of Meningiomas by Weighted Gene Coexpression Network Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4927547. [PMID: 32596316 PMCID: PMC7303753 DOI: 10.1155/2020/4927547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/28/2020] [Accepted: 05/09/2020] [Indexed: 02/04/2023]
Abstract
Meningiomas are the most common primary intracranial tumor in adults. However, to date, systemic coexpression analyses for meningiomas fail to explain its pathogenesis. The aim of the present study was to construct coexpression modules and identify potential biomarkers associated with meningioma progression. Weighted gene coexpression network analysis (WGCNA) was performed based on GSE43290, and module preservation was tested by GSE74385. Functional annotations were performed to analyze biological significance. Hub genes were selected for efficacy evaluations and correlation analyses using two independent cohorts. A total of 14 coexpression modules were identified, and module lightcyan was significantly associated with WHO grades. Functional enrichment analyses of module lightcyan were associated with tumor pathogenesis. The top 10 hub genes were extracted. Ten biomarkers, particularly AHCYL2, FGL2, and KCNMA1, were significantly related to grades and prognosis of meningioma. These findings not only construct coexpression modules leading to the better understanding of its pathogenesis but also provide potential biomarkers that represent specific on tumor grades and identify recurrence, predicting prognosis and progression of meningiomas.
Collapse
|