1
|
Loda A, Calza S, Giacomini A, Ravelli C, Krishna Chandran AM, Tobia C, Tabellini G, Parolini S, Semeraro F, Ronca R, Rezzola S. FGF-trapping hampers cancer stem-like cells in uveal melanoma. Cancer Cell Int 2023; 23:89. [PMID: 37165394 PMCID: PMC10173517 DOI: 10.1186/s12935-023-02903-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/24/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Cancer stem-like cells (CSCs) are a subpopulation of tumor cells responsible for tumor initiation, metastasis, chemoresistance, and relapse. Recently, CSCs have been identified in Uveal Melanoma (UM), which represents the most common primary tumor of the eye. UM is highly resistant to systemic chemotherapy and effective therapies aimed at improving overall survival of patients are eagerly required. METHODS Herein, taking advantage from a pan Fibroblast Growth Factor (FGF)-trap molecule, we singled out and analyzed a UM-CSC subset with marked stem-like properties. A hierarchical clustering of gene expression data publicly available on The Cancer Genome Atlas (TCGA) was performed to identify patients' clusters. RESULTS By disrupting the FGF/FGF receptor (FGFR)-mediated signaling, we unmasked an FGF-sensitive UM population characterized by increased expression of numerous stemness-related transcription factors, enhanced aldehyde dehydrogenase (ALDH) activity, and tumor-sphere formation capacity. Moreover, FGF inhibition deeply affected UM-CSC survival in vivo in a chorioallantoic membrane (CAM) tumor graft assay, resulting in the reduction of tumor growth. At clinical level, hierarchical clustering of TCGA gene expression data revealed a strong correlation between FGFs/FGFRs and stemness-related genes, allowing the identification of three distinct clusters characterized by different clinical outcomes. CONCLUSIONS Our findings support the evidence that the FGF/FGFR axis represents a master regulator of cancer stemness in primary UM tumors and point to anti-FGF treatments as a novel therapeutic strategy to hit the CSC component in UM.
Collapse
Affiliation(s)
- Alessandra Loda
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Stefano Calza
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Adwaid Manu Krishna Chandran
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Chiara Tobia
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Giovanna Tabellini
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Silvia Parolini
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy
| | - Francesco Semeraro
- Eye Clinic, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy.
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, viale Europa 11, 25123, Brescia, Italy.
| |
Collapse
|
2
|
Fan Z, Duan J, Luo P, Shao L, Chen Q, Tan X, Zhang L, Xu X. SLC25A38 as a novel biomarker for metastasis and clinical outcome in uveal melanoma. Cell Death Dis 2022; 13:330. [PMID: 35411037 PMCID: PMC9001737 DOI: 10.1038/s41419-022-04718-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 02/18/2022] [Accepted: 03/09/2022] [Indexed: 01/03/2023]
Abstract
Risk of metastasis is increased by the presence of chromosome 3 monosomy in uveal melanoma (UM). This study aimed to identify more accurate biomarker for risk of metastasis in UM. A total of 80 patients with UM from TCGA were assigned to two groups based on the metastatic status, and bioinformatic analyses were performed to search for critical genes for risk of metastasis. SLC25A38, located on chromosome 3, was the dominant downregulated gene in metastatic UM patients. Low expression of SLC25A38 was an independent predictive and prognostic factor in UM. The predictive potential of SLC25A38 expression was superior to that of pervious reported biomarkers in both TCGA cohort and GSE22138 cohort. Subsequently, its role in promoting metastasis was explored in vitro and in vivo. Knock-out of SLC25A38 could enhance the migration ability of UM cells, and promote distant metastasis in mice models. Through the inhibition of CBP/HIF-mediated pathway followed by the suppression of pro-angiogenic factors, SLC25A38 was situated upstream of metastasis-related pathways, especially angiogenesis. Low expression of SLC25A38 promotes angiogenesis and metastasis, and identifies increased metastatic risk and worse survival in UM patients. This finding may further improve the accuracy of prognostic prediction for UM.
Collapse
Affiliation(s)
- Zhongyi Fan
- Department of Oncology and Bio-therapeutic Center, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen Research Center for Communicable Disease Diagnosis and Treatment, Shenzhen, 518112, China.,Department of Oncology, The First Medical Center, General Hospital of PLA, Beijing, 100853, China
| | - Jingjing Duan
- Department of Gastrointestinal Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Pu Luo
- Department of Oncology and Bio-therapeutic Center, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen Research Center for Communicable Disease Diagnosis and Treatment, Shenzhen, 518112, China
| | - Ling Shao
- Department of Oncology and Bio-therapeutic Center, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen Research Center for Communicable Disease Diagnosis and Treatment, Shenzhen, 518112, China
| | - Qiong Chen
- Department of Oncology and Bio-therapeutic Center, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen Research Center for Communicable Disease Diagnosis and Treatment, Shenzhen, 518112, China
| | - Xiaohua Tan
- Department of Oncology and Bio-therapeutic Center, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen Research Center for Communicable Disease Diagnosis and Treatment, Shenzhen, 518112, China.
| | - Lei Zhang
- Department of Ophthalmology, Xuanwu Hospital Attached to the Capital Medical University, Beijing, 100053, China.
| | - Xiaojie Xu
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, 100850, China.
| |
Collapse
|
3
|
Exploring the FGF/FGFR System in Ocular Tumors: New Insights and Perspectives. Int J Mol Sci 2022; 23:ijms23073835. [PMID: 35409195 PMCID: PMC8998873 DOI: 10.3390/ijms23073835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/16/2022] Open
Abstract
Ocular tumors are a family of rare neoplasms that develop in the eye. Depending on the type of cancer, they mainly originate from cells localized within the retina, the uvea, or the vitreous. Even though current treatments (e.g., radiotherapy, transpupillary thermotherapy, cryotherapy, chemotherapy, local resection, or enucleation) achieve the control of the local tumor in the majority of treated cases, a significant percentage of patients develop metastatic disease. In recent years, new targeting therapies and immuno-therapeutic approaches have been evaluated. Nevertheless, the search for novel targets and players is eagerly required to prevent and control tumor growth and metastasis dissemination. The fibroblast growth factor (FGF)/FGF receptor (FGFR) system consists of a family of proteins involved in a variety of physiological and pathological processes, including cancer. Indeed, tumor and stroma activation of the FGF/FGFR system plays a relevant role in tumor growth, invasion, and resistance, as well as in angiogenesis and dissemination. To date, scattered pieces of literature report that FGFs and FGFRs are expressed by a significant subset of primary eye cancers, where they play relevant and pleiotropic roles. In this review, we provide an up-to-date description of the relevant roles played by the FGF/FGFR system in ocular tumors and speculate on its possible prognostic and therapeutic exploitation.
Collapse
|
4
|
Meng Z, Chen Y, Wu W, Yan B, Zhang L, Chen H, Meng Y, Liang Y, Yao X, Luo J. PRRX1 Is a Novel Prognostic Biomarker and Facilitates Tumor Progression Through Epithelial–Mesenchymal Transition in Uveal Melanoma. Front Immunol 2022; 13:754645. [PMID: 35281030 PMCID: PMC8914230 DOI: 10.3389/fimmu.2022.754645] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/31/2022] [Indexed: 01/10/2023] Open
Abstract
Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. UM develops and is sustained by inflammation and immunosuppression from the tumor microenvironment (TME). This study sought to identify a reliable TME-related biomarker that could provide survival prediction and new insight into therapy for UM patients. Based on clinical characteristics and the RNA-seq transcriptome data of 80 samples from The Cancer Genome Atlas (TCGA) database, PRRX1 as a TME- and prognosis-related gene was identified using the ESTIMATE algorithm and the LASSO–Cox regression model. A prognostic model based on PRRX1 was constructed and validated with a Gene Expression Omnibus (GEO) dataset of 63 samples. High PRRX1 expression was associated with poorer overall survival (OS) and metastasis-free survival (MFS) in UM patients. Comprehensive results of the prognostic analysis showed that PRRX1 was an independent and reliable predictor of UM. Then the results of immunological characteristics demonstrated that higher expression of PRRX1 was accompanied by higher expression of immune checkpoint genes, lower tumor mutation burden (TMB), and greater tumor cell infiltration into the TME. Gene set enrichment analysis (GSEA) showed that high PRRX1 expression correlated with angiogenesis, epithelial–mesenchymal transition (EMT), and inflammation. Furthermore, downregulation of PRRX1 weakened the process of EMT, reduced cell invasion and migration of human UM cell line MuM-2B in vitro. Taken together, these findings indicated that increased PRRX1 expression is independently a prognostic factor of poorer OS and MFS in patients with UM, and that PRRX1 promotes malignant progression of UM by facilitating EMT, suggesting that PRRX1 may be a potential target for UM therapy.
Collapse
Affiliation(s)
- Zhishang Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yanzhu Chen
- Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wenyi Wu
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Yan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lusi Zhang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Huihui Chen
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yongan Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Youling Liang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxi Yao
- Shenzhen College of International Education, Shenzhen, China
| | - Jing Luo
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Jing Luo,
| |
Collapse
|
5
|
Wang J, Zhao H, Zheng L, Zhou Y, Wu L, Xu Y, Zhang X, Yan G, Sheng H, Xin R, Jiang L, Lei J, Zhang J, Chen Y, Peng J, Chen Q, Yang S, Yu K, Li D, Xie Q, Li Y. FGF19/SOCE/NFATc2 signaling circuit facilitates the self-renewal of liver cancer stem cells. Am J Cancer Res 2021; 11:5045-5060. [PMID: 33754043 PMCID: PMC7978301 DOI: 10.7150/thno.56369] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/31/2021] [Indexed: 02/07/2023] Open
Abstract
Background & Aims: Liver cancer stem cells (LCSCs) mediate therapeutic resistance and correlate with poor outcomes in patients with hepatocellular carcinoma (HCC). Fibroblast growth factor (FGF)-19 is a crucial oncogenic driver gene in HCC and correlates with poor prognosis. However, whether FGF19 signaling regulates the self-renewal of LCSCs is unknown. Methods: LCSCs were enriched by serum-free suspension. Self-renewal of LCSCs were characterized by sphere formation assay, clonogenicity assay, sorafenib resistance assay and tumorigenic potential assays. Ca2+ image was employed to determine the intracellular concentration of Ca2+. Gain- and loss-of function studies were applied to explore the role of FGF19 signaling in the self-renewal of LCSCs. Results: FGF19 was up-regulated in LCSCs, and positively correlated with certain self-renewal related genes in HCC. Silencing FGF19 suppressed self-renewal of LCSCs, whereas overexpressing FGF19 facilitated CSCs-like properties via activation of FGF receptor (FGFR)-4 in none-LCSCs. Mechanistically, FGF19/FGFR4 signaling stimulated store-operated Ca2+ entry (SOCE) through both the PLCγ and ERK1/2 pathways. Subsequently, SOCE-calcineurin signaling promoted the activation and translocation of nuclear factors of activated T cells (NFAT)-c2, which transcriptionally activated the expression of stemness-related genes (e.g., NANOG, OCT4 and SOX2), as well as FGF19. Furthermore, blockade of FGF19/FGFR4-NFATc2 signaling observably suppressed the self-renewal of LCSCs. Conclusions: FGF19/FGFR4 axis promotes the self-renewal of LCSCs via activating SOCE/NFATc2 pathway; in turn, NFATc2 transcriptionally activates FGF19 expression. Targeting this signaling circuit represents a potential strategy for improving the therapeutic efficacy of HCC.
Collapse
|
6
|
Cancer stem cell transcriptome landscape reveals biomarkers driving breast carcinoma heterogeneity. Breast Cancer Res Treat 2021; 186:89-98. [PMID: 33389402 DOI: 10.1007/s10549-020-06045-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/02/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Breast carcinomas are heterogeneous diseases with distinct clinical outcomes and cancer stem cell (CSC) percentages. Exploring breast carcinoma stem cell landscape could help understand the heterogeneity of such cancers with profound clinical relevance. METHODS We conducted transcriptional profiling of CSCs and non-stem cancer cells isolated from three triple-negative breast carcinoma cell lines, analyzed the CSC transcriptome landscape that drives breast carcinoma heterogeneity through differentially expressed gene identification, gene ontology (GO) and pathway enrichment analyses as well as network construction, and experimentally validated the network hub gene. RESULTS We identified a CSC feature panel consisting of 122 and 381 over-represented and under-expressed genes capable of differentiating breast carcinoma subtypes. We also underpinned the prominent roles of the PI3K-AKT pathway in empowering carcinoma cells with uncontrolled proliferative and migrative abilities that ultimately foster cancer stemness, and revealed the potential promotive roles of ATP6V1B1 on breast carcinoma stemness through functional in vitro studies. CONCLUSIONS Our study contributes in identifying a CSC feature panel for breast carcinomas that drives breast carcinoma heterogeneity at the transcriptional level, which provides a reservoir for diagnostic marker and/or therapeutic target identification once experimentally validated as demonstrated by ATP6V1B1.
Collapse
|
7
|
Guo Y, Zhu J, Wang X, Li R, Jiang K, Chen S, Fan J, Xue L, Hao D. Orai1 Promotes Osteosarcoma Metastasis by Activating the Ras-Rac1-WAVE2 Signaling Pathway. Med Sci Monit 2019; 25:9227-9236. [PMID: 31796725 PMCID: PMC6909920 DOI: 10.12659/msm.919594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background The purpose of this study was to investigate whether Orai1 plays a role in the metastasis of osteosarcoma. Material/Methods The expression of Orai1 was silenced by small interfering RNAs against Orai1 (Orai1 siRNA) in osteosarcoma MG-63 cells. Various experiments were carried out to detect the changes in migration, invasion, and adhesion ability of these osteosarcoma cells. Furthermore, the activity of Rac1, Wave2, and Ras was detected using Western blot analysis. Moreover, the Rac1 and Ras inhibitors were used to confirm whether the Ras-Rac1-WAVE2 signaling pathway was involved in osteosarcoma metastasis promoted by Orai1. Results We found that the migration, invasion, and adhesion ability of MG-63 cells were significantly reduced after silencing Orai1 expression (p<0.05). Moreover, the activity of the Rac1-WAVE2 signaling pathway was significantly inhibited after silencing of Orai1 expression (p<0.05). After the Rac1 inhibitor was added, Orai1 siRNA could not further inhibit migration, invasion, and adhesion of the osteosarcoma cells. Further experiments showed that Ras activity was significantly inhibited after silencing Orai1 expression (p<0.05). Moreover, Orai1 siRNA did not further inhibit the activity of the Rac1-WAVE2 signaling pathway nor did it further inhibit the migration, invasion, and adhesion ability of osteosarcoma cells following the addition of Ras inhibitors. Conclusions Orai1 activates the Ras-Rac1-WAVE2 signaling pathway to promote metastasis of osteosarcoma. Abnormal expression or function of Orai1 may be an important cause of osteosarcoma metastasis.
Collapse
Affiliation(s)
- Yunshan Guo
- Department of Spinal Surgery, Hong Hui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Jinwen Zhu
- Department of Spinal Surgery, Hong Hui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Xiaodong Wang
- Department of Spinal Surgery, Hong Hui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Ruoyu Li
- Department of Spinal Surgery, Hong Hui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Kuo Jiang
- Department of Spinal Surgery, Hong Hui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Shi Chen
- Department of Emergency Medicine, Hong Hui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Jinzhu Fan
- Department of Orthopedics, Hong Hui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Liujie Xue
- Department of Spinal Surgery, Hong Hui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Dingjun Hao
- Department of Spinal Surgery, Hong Hui Hospital, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
8
|
Li L, Zhang S, Li H, Chou H. FGFR3 promotes the growth and malignancy of melanoma by influencing EMT and the phosphorylation of ERK, AKT, and EGFR. BMC Cancer 2019; 19:963. [PMID: 31619201 PMCID: PMC6796326 DOI: 10.1186/s12885-019-6161-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 09/13/2019] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Overexpression of fibroblast growth factor receptor 3 (FGFR3) has been linked to tumor progression in many types of cancer. The role of FGFR3 in melanoma remains unclear. In this study, we aimed to uncover the role of FGFR3 in the growth and metastasis of melanoma. METHODS FGFR3 knockdown and overexpression strategies were employed to investigate the effects of FGFR3 on colony formation, cell apoptosis, proliferation, migration, and in vitro invasion, along with the growth and metastasis of melanoma in a xenografts mouse model. The protein expression levels of extracellular signal-regulated kinase (ERK), protein kinase B (AKT), epidermal growth factor receptor (EGFR), and epithelial-mesenchymal transition (EMT) markers were determined by Western blot analysis. RESULTS The mRNA expression of FGFR3 was higher in melanoma tissues than normal healthy tissues. FGFR3 expression in cutaneous malignant melanoma (CMM) tissues was positively correlated with the Breslow thickness and lymph node metastasis. In A357 cells, knockdown of the FGFR3 gene decreased the colony formation ability, cell proliferation, invasion, and migration, but increased the caspase 3 activity and the apoptosis rate; overexpression of FGFR3 increased the colony formation ability, cell proliferation, invasion, and migration, but decreased the caspase 3 activity and apoptosis rates. FGFR3 knockdown also upregulated E-cadherin, downregulated N-cadherin and vimentin, and decreased the phosphorylation levels of ERK, AKT, and EGFR. In the MCC xenografts mice, knockdown of FGFR3 decreased tumor growth and metastasis. CONCLUSIONS FGFR3, which is highly expressed in CMM tissues, is correlated with increased Breslow thickness and lymph node metastasis. FGFR3 promotes melanoma growth, metastasis, and EMT behaviors, likely by affecting the phosphorylation levels of ERK, AKT, and EGFR.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cadherins/metabolism
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation/genetics
- Epithelial-Mesenchymal Transition/genetics
- ErbB Receptors/metabolism
- Heterografts
- Humans
- MAP Kinase Signaling System
- Male
- Melanoma/metabolism
- Melanoma/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Invasiveness/genetics
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Transfection
- Vimentin/metabolism
- Melanoma, Cutaneous Malignant
Collapse
Affiliation(s)
- Lei Li
- Department of Plastic and Cosmetic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, 450003 Henan China
| | - Shuai Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Hao Li
- Department of Plastic and Cosmetic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, 450003 Henan China
| | - Haiyan Chou
- Department of Plastic and Cosmetic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, 450003 Henan China
| |
Collapse
|
9
|
The Autocrine FGF/FGFR System in both Skin and Uveal Melanoma: FGF Trapping as a Possible Therapeutic Approach. Cancers (Basel) 2019; 11:cancers11091305. [PMID: 31487962 PMCID: PMC6770058 DOI: 10.3390/cancers11091305] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022] Open
Abstract
Fibroblast growth factors (FGFs) play non-redundant autocrine/paracrine functions in various human cancers. The Cancer Genome Atlas (TCGA) data mining indicates that high levels of FGF and/or FGF receptor (FGFR) expression are associated with reduced overall survival, chromosome 3 monosomy and BAP1 mutation in human uveal melanoma (UM), pointing to the FGF/FGFR system as a target for UM treatment. Here, we investigated the impact of different FGF trapping approaches on the tumorigenic and liver metastatic activity of liver metastasis-derived murine melanoma B16-LS9 cells that, similar to human UM, are characterized by a distinctive hepatic tropism. In vitro and in vivo experiments demonstrated that the overexpression of the natural FGF trap inhibitor long-pentraxin 3 (PTX3) inhibits the oncogenic activity of B16-LS9 cells. In addition, B16-LS9 cells showed a reduced tumor growth and liver metastatic activity when grafted in PTX3-overexpressing transgenic mice. The efficacy of the FGF trapping approach was confirmed by the capacity of the PTX3-derived pan-FGF trap small molecule NSC12 to inhibit B16-LS9 cell growth in vitro, in a zebrafish embryo orthotopic tumor model and in an experimental model of liver metastasis. Possible translational implications for these observations were provided by the capacity of NSC12 to inhibit FGF signaling and cell proliferation in human UM Mel285, Mel270, 92.1, and OMM2.3 cells. In addition, NSC12 caused caspase-3 activation and PARP cleavage followed by apoptotic cell death as well as β-catenin degradation and inhibition of UM cell migration. Together, our findings indicate that FGF trapping may represent a novel therapeutic strategy in UM.
Collapse
|