1
|
Wang Z, Muthusamy V, Petrylak DP, Anderson KS. Tackling FGFR3-driven bladder cancer with a promising synergistic FGFR/HDAC targeted therapy. NPJ Precis Oncol 2023; 7:70. [PMID: 37479885 PMCID: PMC10362036 DOI: 10.1038/s41698-023-00417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 06/23/2023] [Indexed: 07/23/2023] Open
Abstract
Bladder cancer (BC) is one of the most prevalent malignancies worldwide and FGFR3 alterations are particularly common in BC. Despite approval of erdafitinib, durable responses for FGFR inhibitors are still uncommon and most patients relapse to metastatic disease. Given the necessity to discover more efficient therapies for BC, herein, we sought to explore promising synergistic combinations for BC with FGFR3 fusions. Our studies confirmed the synergy between FGFR and HDAC inhibitors in vitro and demonstrated its benefits in vivo. Mechanistic studies revealed that quisinostat can downregulate FGFR3 expression by suppressing FGFR3 translation. Additionally, quisinostat can also sensitize BC cells to erdafitinib by downregulating HDGF. Furthermore, the synergy was also confirmed in BC cells with FGFR3 S249C. This study discovers a new avenue for treatment of FGFR3-driven BC and uncovers new mechanistic insights. These preclinical studies pave the way for a direct translation of this combination to early phase clinical trials.
Collapse
Affiliation(s)
- Zechen Wang
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
| | | | | | - Karen S Anderson
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
| |
Collapse
|
2
|
Wang Y, Ma Q, Huo Z. Identification of hub genes, pathways, and related transcription factors in systemic lupus erythematosus: A preliminary bioinformatics analysis. Medicine (Baltimore) 2021; 100:e26499. [PMID: 34160465 PMCID: PMC8238284 DOI: 10.1097/md.0000000000026499] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 05/31/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multiple organ damage and the production of a variety of autoantibodies. The pathogenesis of SLE has not been fully defined, and it is difficult to treat. Our study aimed to identify candidate genes that may be used as biomarkers for the screening, diagnosis, and treatment of SLE. METHODS We used the GEO2R tool to identify the differentially expressed genes (DEGs) in SLE-related datasets retrieved from the Gene Expression Omnibus (GEO). In addition, we also identified the biological functions of the DEGs by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. Additionally, we constructed protein-protein interaction (PPI) networks to identify hub genes, as well as the regulatory network of transcription factors related to DEGs. RESULTS Two datasets were identified for use from the GEO (GSE50772, GSE4588), and 34 up-regulated genes and 4 down-regulated genes were identified by GEO2R. Pathway analysis of the DEGs revealed enrichment of the interferon alpha/beta signaling pathway; GO analysis was mainly enriched in response to interferon alpha, regulation of ribonuclease activity. PPIs were constructed through the STRING database and 14 hub genes were selected and 1 significant module (score = 12.923) was obtained from the PPI network. Additionally, 11 key transcription factors that interacted closely with the 14 hub DEGs were identified from the gene transcription factor network. CONCLUSIONS Bioinformatic analysis is an effective tool for screening the original genomic data in the GEO database, and a large number of SLE-related DEGs were identified. The identified hub DEGs may be potential biomarkers of SLE.
Collapse
|
3
|
Cheng C, Li W, Peng X, Liu X, Zhang Z, Liu Z, Deng T, Luo R, Fang W, Deng X. miR-1254 induced by NESG1 inactivates HDGF/DDX5-stimulated nuclear translocation of β-catenin and suppresses NPC metastasis. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:615-624. [PMID: 33718512 PMCID: PMC7907678 DOI: 10.1016/j.omtm.2021.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 02/01/2021] [Indexed: 12/19/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is one of the most common malignant tumors in Chinese and other Southeast Asians. We aimed to explore the precise mechanism for NESG1 in NPC for understanding the pathogenesis of NPC. Transwell, Boyden assays, and wounding healing were respectively performed for cell metastasis. The microRNA (miRNA) microarray and luciferase reporter assays were designed to clarify NESG1-modulated miRNAs and miR-1254-targeted protein. Western blotting assays examined the pathways regulated by miR-1254, the (Hepatoma-Derived Growth Factor) HDGF/DDX5 complex, and NESG1. The chromatin immunoprecipitation (ChIP), electrophoretic mobility shift assay (EMSA), and co-immunoprecipitation (coIP) assays were used to explore the DNA-protein complex and protein-protein complex. NESG1 suppressed NPC migration and invasion via Wnt/β-catenin signaling. Further, miR-1254 was confirmed as a positive downstream modulator of NESG1 reducing metastatic abilities of NPC cells in vivo and in vitro. Transduction of HDGF significantly restored cell migration and invasion ability in miR-1254-overexpressing NPC cells. In clinical samples, miR-1254 expression was negatively correlated with HDGF and positively correlated with NESG1 expression. miR-1254 acts as an independent prognostic factor for NPC, which was induced by NESG1 to suppress NPC metastasis via inactivating Wnt/β-catenin pathway and its downstream EMT signals.
Collapse
Affiliation(s)
- Chao Cheng
- Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Wenmin Li
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xuemei Peng
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiong Liu
- E.N.T. Department of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ziyan Zhang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhen Liu
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Tongyuan Deng
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Rongcheng Luo
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Weiyi Fang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaojie Deng
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Yin R, Liu J, Zhao D, Wang F. Long Non-Coding RNA ASB16-AS1 Functions as a miR-760 Sponge to Facilitate the Malignant Phenotype of Osteosarcoma by Increasing HDGF Expression. Onco Targets Ther 2020; 13:2261-2274. [PMID: 32214826 PMCID: PMC7081065 DOI: 10.2147/ott.s240022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/27/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose ASB16 antisense RNA 1 (ASB16-AS1) is a cancer-associated long non-coding RNA that contributes to tumorigenesis and tumor development. Nevertheless, to the best of our knowledge, whether and how ASB16-AS1 is implicated in osteosarcoma (OS) malignancy remains unclear and therefore warrants exploration. Our current study focused on making in-depth investigation of ASB16-AS1 in OS. In the present study, the expression pattern of ASB16-AS1 in OS tissues and cell lines was analyzed. In addition, we examined the clinical value of ASB16-AS1 for OS patients. Furthermore, we explored the impacts of ASB16-AS1 on the malignant phenotype of OS cells in vitro and in vivo as well as the underlying mechanism. Methods ASB16-AS1, microRNA-760 (miR-760) and hepatoma-derived growth factor (HDGF) expressions were measured using reverse transcription-quantitative PCR. Cell proliferation and apoptosis were evaluated using CCK-8 and flow cytometry analyses, respectively, and cell migration and invasion were determined via cell migration and invasion assays. Results ASB16-AS1 expression was significantly elevated in OS tissues and cell lines, and increased ASB16-AS1 expression was related to patients' tumor size, TNM stage, and distant metastasis. The overall survival rate of OS patients presenting high ASB16-AS1 expression was shorter than that of patients presenting low ASB16-AS1 expression. Reduced ASB16-AS1 expression inhibited OS cell proliferation, migration, and invasion; promoted cell apoptosis; and impaired tumor growth in vivo. Mechanistically, ASB16-AS1 served as a sponge for miR-760 and positively modulated the expression of its target HDGF. Finally, inhibiting miR-760 and restoring HDGF expression abolished the impacts of ASB16-AS1 knockdown on the malignant characteristics of OS cells. Conclusion ASB16-AS1 is a novel oncogenic lncRNA in OS cells. ASB16-AS1 increased HDGF expression by sponging miR-760, thereby conferring cancer-promoting roles in OS. ASB16-AS1 is a potential early diagnostic and therapeutic target in OS.
Collapse
Affiliation(s)
- Ruofeng Yin
- Department of Orthopedics, China-Japan Union Hospital Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Junzhi Liu
- Department of Quality Control, China-Japan Union Hospital Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Dongxu Zhao
- Department of Orthopedics, China-Japan Union Hospital Jilin University, Changchun, Jilin 130033, People's Republic of China
| | - Fei Wang
- Department of Orthopedics, China-Japan Union Hospital Jilin University, Changchun, Jilin 130033, People's Republic of China
| |
Collapse
|
5
|
Zhang C, Chang X, Chen D, Yang F, Li Z, Li D, Yu N, Yan L, Liu H, Xu Z. Downregulation of HDGF inhibits the tumorigenesis of bladder cancer cells by inactivating the PI3K-AKT signaling pathway. Cancer Manag Res 2019; 11:7909-7923. [PMID: 31692549 PMCID: PMC6710542 DOI: 10.2147/cmar.s215341] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 06/28/2019] [Indexed: 12/29/2022] Open
Abstract
Background Hepatoma-derived growth factor (HDGF) is a heparin-binding protein that has been observed to be abnormally expressed in numerous malignancies, but the definite role of HDGF in bladder cancer (BCa) has not been clarified. Here, we conduct the present study to evaluate correlations between HDGF and BCa. Methods Bioinformatics analysis was used to evaluate HDGF expression levels in BCa tissues. The effect of HDGF on cell proliferation, migration, invasion, cell cycle and apoptosis was analyzed utilizing CCK-8, clone formation, Transwell assays and flow cytometry, respectively. In addition, the xenograft tumor model was established. Results Based on bioinformatics analysis, we noticed that HDGF was highly expressed in BCa tissues and was positively correlated with poor prognosis in patients. Knockdown of HDGF markedly reduced tumorigenesis in BCa cells. Furthermore, the results of flow cytometry showed that HDGF deletion enhanced apoptosis in T24 and 253J cells and led to cell cycle arrest in G1 phase. In further studies, we found that tumor growth was inhibited in xenograft nude mouse models with HDGF deletion. The results of RNA-seq analysis revealed that the PI3K-AKT signaling pathway-related genes were obviously changed in HDGF-deficient 253J cells, and this result was further confirmed by Western blot analysis. Conclusion In summary, we suggest that HDGF plays a substantial role in BCa and promotes tumor development and progression by regulating the PI3K-AKT signaling pathway, which provides a promising target for BCa treatment.
Collapse
Affiliation(s)
- Cong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Shandong University, Jinan, People's Republic of China
| | - Xiangping Chang
- Department of Urology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Dongshan Chen
- Department of Urology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Feilong Yang
- Department of Urology, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Zeyan Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Dawei Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Nengwang Yu
- Department of Urology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Lei Yan
- Department of Urology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Hainan Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| | - Zhonghua Xu
- Department of Urology, Qilu Hospital of Shandong University, Jinan 250012, People's Republic of China
| |
Collapse
|
6
|
Zhao C, Li H, Wang L, Sun W. An Immunohistochemical Study of Stathmin 1 Expression in Osteosarcoma Shows an Association with Metastases and Poor Patient Prognosis. Med Sci Monit 2018; 24:6070-6078. [PMID: 30169496 PMCID: PMC6129035 DOI: 10.12659/msm.910953] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Osteosarcoma is the most common primary bone cancer and has a broad spectrum of histological subtypes. Stathmin 1 (STMN1) is a cytosolic phosphoprotein that is expressed in several types of cancer. The aim of this study was to evaluate the expression levels of STMN1 in osteosarcoma with clinicopathological characteristics and patient prognosis. Material/Methods The expression of STMN1 in tumor tissue from 94 patients with OS was detected and evaluated using an immunohistochemical score to divide the patients into low expression and high expression groups. Correlation between STMN1 expression and clinicopathological factors were analyzed with Fisher’s test, the prognostic value of expression levels of STMN1 in tumor tissue was evaluated by Kaplan-Meier univariate analysis, and independent prognostic factors were identified using the Cox regression model. Results Low expression of STMN1 was found in 43.62% of cases and high expression of STMN1 was found in 56.38% of cases of osteosarcoma. High tumor expression of STMN1 was significantly associated with the presence of metastases (P=0.028), Enneking surgical stage (P=0.030), tumor response to chemotherapy (P=0.011), and the site of tumor origin (P=0.023). High tumor expression of STMN1 was a prognostic marker in patients with osteosarcoma for poor prognosis (P=0.016), poor response to chemotherapy (P=0.004), the presence of metastases (P=0.003), advanced Enneking surgical stage (P=0.014), and the chondroblastic osteosarcoma subtype (P=0.004). The expression STMN1 was identified as an independent prognostic biomarker of osteosarcoma. Conclusions Increased expression of STMN1 in tumor tissue was an independent prognostic biomarker in patients with osteosarcoma.
Collapse
Affiliation(s)
- Changlei Zhao
- Department of Rehabilitation, Yidu Central Hospital, Weifang, Shandong, China (mainland)
| | - Hailing Li
- Department of Rehabilitation, Yidu Central Hospital, Weifang, Shandong, China (mainland)
| | - Lingling Wang
- Department of Geriatrics, Nursing Vocational College of Weifang, Weifang, Shandong, China (mainland)
| | - Wei Sun
- Department of Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China (mainland)
| |
Collapse
|
7
|
Min X, Wen J, Zhao L, Wang K, Li Q, Huang G, Liu J, Zhao X. Role of hepatoma-derived growth factor in promoting de novo lipogenesis and tumorigenesis in hepatocellular carcinoma. Mol Oncol 2018; 12:1480-1497. [PMID: 30004626 PMCID: PMC6120245 DOI: 10.1002/1878-0261.12357] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/11/2018] [Accepted: 06/26/2018] [Indexed: 01/05/2023] Open
Abstract
Although identified as a growth factor, the mechanism by which hepatoma‐derived growth factor (HDGF) promotes cancer development remains unclear. We found that nuclear but not cytoplasmic HDGF is closely associated with prognosis of hepatocellular carcinoma (HCC). RNA‐sequencing analysis further demonstrated that the nuclear role of HDGF involved regulation of transcription of lipid metabolism genes. HDGF‐induced expression of lipogenic genes was mainly associated with activation of sterol regulatory element binding protein (SREBP) transcription factor. Coexpression of SREBP‐1 and nuclear HDGF predicts poor prognosis for HCC. In addition, by changing the first amino acid of the PWWP domain from proline to alanine, the type of PWWP domain changed from P‐ to A‐type, resulting in inability to induce SREBP‐1‐mediated gene transcription. The type of PWWP domain affects the recruitment of the C‐terminal binding protein‐1 transcriptional repressor on the promoter of the lipogenic gene. Our data indicate that HDGF acts as a coactivator of SREBP1‐mediated transcription of lipogenic genes. The PWWP domain is crucial for HDGF to promote lipogenesis. Moreover, transcriptional regulation of nuclear HDGF plays important roles in the development of HCC.
Collapse
Affiliation(s)
- Xuejie Min
- Department of Nuclear Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jun Wen
- Department of Nuclear Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Li Zhao
- Department of Nuclear Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Kaiying Wang
- Department of Nuclear Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Qingli Li
- Department of Nuclear Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Gang Huang
- Shanghai University of Medicine & Health Sciences, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xiaoping Zhao
- Department of Nuclear Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| |
Collapse
|