1
|
Wang J, Du J, Luo X, Guo L, Liu Y, Zhou J, Zou Y, Lu Z, Pan X, Chen X, Zhong A, Wan X, Wang L, Liu H, Dai S, Zhang S, Xiong X, Tan P, Wang M, Wu B, Zhang Q, Wang Y, Zhang M, Lu R, Lin H, Li Y, Li Y, Han Z, Chen L, Hu B, Liu Y, Na F, Chen C. A platform of functional studies of ESCC-associated gene mutations identifies the roles of TGFBR2 in ESCC progression and metastasis. Cell Rep 2024; 43:114952. [PMID: 39527477 DOI: 10.1016/j.celrep.2024.114952] [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: 03/11/2024] [Revised: 08/31/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Genomics studies have detected numerous genetic alterations in esophageal squamous cell carcinoma (ESCC). However, the functions of these mutations largely remain elusive, partially due to a lack of feasible animal models. Here, we report a convenient platform with CRISPR-Cas9-mediated introduction of genetic alterations and orthotopic transplantation to generate a series of primary ESCC models in mice. With this platform, we validate multiple frequently mutated genes, including EP300, FAT1/2/4, KMT2D, NOTCH2, and TGFBR2, as tumor-suppressor genes in ESCC. Among them, TGFBR2 loss dramatically promotes tumorigenesis and multi-organ metastasis. Paradoxically, TGFBR2 deficiency leads to Smad3 activation, and disruption of Smad3 partially restrains the progression of Tgfbr2-mutated tumors. Drug screening with tumor organoids identifies that pinaverium bromide represses Smad3 activity and restrains Tgfbr2-deficient ESCC. Our studies provide a highly efficient platform to investigate the in vivo functions of ESCC-associated mutations and develop potential treatments for this miserable malignancy.
Collapse
Affiliation(s)
- Jian Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiajia Du
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiangmeng Luo
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linjie Guo
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yixin Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianfeng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yang Zou
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenghao Lu
- Chengdu OrganoidMed Medical Laboratory, West China Health Valley, Chengdu, Sichuan 610041, China
| | - Xiangyu Pan
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuelan Chen
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ailing Zhong
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xudong Wan
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongyu Liu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Siqi Dai
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shiyu Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingyu Xiong
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Tan
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Manli Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Baohong Wu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qi Zhang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yingjie Wang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mengsha Zhang
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Runda Lu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huahang Lin
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Li
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yaxin Li
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zongkai Han
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Bing Hu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yu Liu
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Feifei Na
- Department of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Chong Chen
- Department of Gastroenterology, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, No387-201 Hemin st., Chengdu, Sichuan 610212, China; Children's Medicine Key Laboratory of Sichuan Province, Sichuan 610041, China.
| |
Collapse
|
2
|
Wang K, Zhang X, Fan Y, Zhou L, Duan Y, Li S, Sun Z, Zhang C, Yang H, Yuan W, Peng L, Ma X, Xiang S, Wang T, Yang M, Zhang Z, Wang J, Wang Z, Qian M. Reactivation of MAPK-SOX2 pathway confers ferroptosis sensitivity in KRAS G12C inhibitor resistant tumors. Redox Biol 2024; 78:103419. [PMID: 39527862 DOI: 10.1016/j.redox.2024.103419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
The clinical success of KRASG12C inhibitors (G12Ci) including AMG510 and MRTX849 is limited by the eventual development of acquired resistance. A novel and effective treatment to revert or target this resistance is urgent. To this end, we established G12Ci (AMG510 and MRTX849) resistant KRASG12C mutant cancer cell lines and screened with an FDA-approved drug library. We found the ferroptosis inducers including sorafenib and lapatinib stood out with an obvious growth inhibition in the G12Ci resistant cells. Mechanistically, the G12Ci resistant cells exhibited reactivation of MAPK signaling, which repressed SOX2-mediated expression of cystine transporter SLC7A11 and iron exporter SLC40A1. Consequently, the low intracellular GSH level but high iron content engendered hypersensitivity of these resistant tumors to ferroptosis inducers. Ectopic overexpression of SOX2 or SLC7A11 and SLC40A1 conferred resistance to ferroptosis in the G12Ci resistant cells. Ferroptosis induced by sulfasalazine (SAS) achieved obvious inhibition on the tumor growth of xenografts derived from AMG510-resistant KRASG12C-mutant cells. Collectively, our results suggest a novel therapeutic strategy to treat patients bearing G12Ci resistant cancers with ferroptosis inducers.
Collapse
Affiliation(s)
- Kai Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Xin Zhang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Yufei Fan
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Liang Zhou
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yajun Duan
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Su Li
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhongkan Sun
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Chunqian Zhang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Haoyu Yang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Wenxiu Yuan
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Linyuan Peng
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoyu Ma
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Siliang Xiang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Tianzhi Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Mei Yang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhenyuan Zhang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiaxuan Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhongyuan Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| | - Minxian Qian
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
3
|
Zhu F, Wang P, Zhang Z, Yao C, Wang Y, Ye J, Wu J. Integrative genomic analysis reveals cancer-associated mutations in patients with ophthalmic tumors. J Int Med Res 2024; 52:3000605241258171. [PMID: 39053449 PMCID: PMC11283671 DOI: 10.1177/03000605241258171] [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: 09/12/2023] [Accepted: 05/04/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE Apart from the role of the retinoblastoma gene, the genomic events associated with poor outcomes in patients with ophthalmic tumors are poorly understood. METHODS We retrospectively analyzed 48 patients with six types of ophthalmic tumors. We searched for high-frequency mutated genes and susceptibility genes in these patients using combined exome and transcriptome analysis. RESULTS We identified four clearly causative genes (TP53, PTCH1, SMO, BAP1). Susceptibility gene analysis identified hotspot genes, including RUNX1, APC, IDH2, and BRCA2, and high-frequency gene analysis identified several genes, including TP53, TTN, and MUC16. Transcriptome analysis identified 5868 differentially expressed genes, of which TOP2A and ZWINT were upregulated in all samples, while CFD, ELANE, HBA1, and HBB were downregulated. Kyoto Encyclopedia of Genes and Genomes enrichment analysis indicated that the phosphoinositide 3-kinase (PI3K)-Akt and Transcriptional misregulation in cancer signaling pathways may be involved in ophthalmic tumorigenesis. CONCLUSIONS TP53 is clearly involved in ophthalmic tumorigenesis, especially in basal cell carcinoma, and the PI3K-Akt signaling pathway may be an essential pathway involved in ophthalmic tumorigenesis. RUNX1, SMO, TOP2A, and ZWINT are also highly likely to be involved in ophthalmic tumorigenesis, but further functional experiments are needed to verify the mechanisms of these genes in regulating tumorigenesis.
Collapse
Affiliation(s)
| | | | | | - Chunlei Yao
- Department of Ophthalmology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yijie Wang
- Department of Ophthalmology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juan Ye
- Department of Ophthalmology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jian Wu
- MyGenostics Inc., Beijing, China
| |
Collapse
|
4
|
Chen Y, Zhang K, Zhang R, Wang Z, Yang L, Zhao T, Zhang S, Lin Y, Zhao H, Liu Y, Wei Y, Zhou Y, Zhang J, Ye X, Zhao J, Li X, Que J, Shi S, Liu K. Targeting the SOX2/CDP protein complex with a peptide suppresses the malignant progression of esophageal squamous cell carcinoma. Cell Death Discov 2023; 9:399. [PMID: 37891174 PMCID: PMC10611744 DOI: 10.1038/s41420-023-01693-7] [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: 05/31/2023] [Revised: 09/28/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Emerging evidence indicates that SOX2 is an oncogene for esophageal squamous cell carcinoma (ESCC). However, direct targeting of SOX2 is not feasible given that this transcription factor plays important roles in the maintenance of tissues such as the brain. Here, we identified CDP (Homeobox protein cut-like 1 or CASP) as a unique SOX2 binding partner enriched in ESCC with Duolink proximity ligation assay, bimolecular fluorescence complementation (BiFc) and immunoprecipitation. We then screened a peptide aptamer library using BiFc and immunoprecipitation and identified several peptide aptamers, including P58, that blocked the CDP/SOX2 interaction, leading to the inhibition of ESCC progress in vitro and in vivo. Upon administration, synthetic peptide P58, containing the YGRKKRRQRRR cell-penetrating peptide and the fluorophore TAMRA, also blocked the growth and metastasis of ESCC in both mice and zebrafish. Therefore, targeting the SOX2 binding partner CDP with peptide P58 offers an alternative avenue to treat ESCC with increased SOX2 levels.
Collapse
Affiliation(s)
- Yunyun Chen
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Kun Zhang
- Department of General Surgery, Fuzhou First General Hospital affiliated with Fujian Medical University, Fuzhou, Fujian, 350009, P. R. China
| | - Rui Zhang
- Department of Laboratory Medicine, The Second Hospital of Fuzhou, Fuzhou, Fujian, 350007, P. R. China
| | - Zhuo Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Liang Yang
- Westlake University, Hangzhou, Zhejiang, 310024, P. R. China
| | - Tingting Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Shihui Zhang
- Centre for Translational Stem Cell Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, 999077, P. R. China
| | - Yong Lin
- Science and Technology Service Center, Fujian Health College, Fuzhou, Fujian, 350101, P. R. China
| | - Hongzhou Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Yongpan Liu
- School of Life Science, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Yuxuan Wei
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Yijian Zhou
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Jiaying Zhang
- School of Life Science, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Xianzong Ye
- Department of Pathology, 900 Hospital of the Joint Logistics Team (Dongfang Hospital, Xiamen University), Fuzhou, Fujian, 350025, P. R. China
| | - Jing Zhao
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Xinxin Li
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Songlin Shi
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China.
| | - Kuancan Liu
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China.
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, P. R. China.
- School of Life Science, Nanchang Normal University, Nanchang, Jiangxi, 330032, P. R. China.
| |
Collapse
|
5
|
Maharati A, Moghbeli M. Long non-coding RNAs as the critical regulators of PI3K/AKT, TGF-β, and MAPK signaling pathways during breast tumor progression. J Transl Med 2023; 21:556. [PMID: 37596669 PMCID: PMC10439650 DOI: 10.1186/s12967-023-04434-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/11/2023] [Indexed: 08/20/2023] Open
Abstract
Breast cancer (BC) as one of the most common causes of human deaths among women, is always considered one of the global health challenges. Despite various advances in diagnostic and therapeutic methods, a significant percentage of BC patients have a poor prognosis due to the lack of therapeutic response. Therefore, investigating the molecular mechanisms involved in BC progression can improve the therapeutic and diagnostic strategies in these patients. Cytokine and growth factor-dependent signaling pathways play a key role during BC progression. In addition to cytokines and growth factors, long non-coding RNAs (lncRNAs) have also important roles in regulation of such signaling pathways. Therefore, in the present review we discussed the role of lncRNAs in regulation of PI3K/AKT, MAPK, and TGF-β signaling pathways in breast tumor cells. It has been shown that lncRNAs mainly have an oncogenic role through the promotion of these signaling pathways in BC. This review can be an effective step in introducing the lncRNAs inhibition as a probable therapeutic strategy to reduce tumor growth by suppression of PI3K/AKT, MAPK, and TGF-β signaling pathways in BC patients. In addition, considering the oncogenic role and increased levels of lncRNAs expressions in majority of the breast tumors, lncRNAs can be also considered as the reliable diagnostic markers in BC patients.
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Fu L, Chen S, He G, Chen Y, Liu B. Targeting Extracellular Signal-Regulated Protein Kinase 1/2 (ERK1/2) in Cancer: An Update on Pharmacological Small-Molecule Inhibitors. J Med Chem 2022; 65:13561-13573. [PMID: 36205714 DOI: 10.1021/acs.jmedchem.2c01244] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Extracellular signal-regulated protein kinase 1/2 (ERK1/2), the only known substrate of MEK1/2, is located downstream of the RAS-RAF-MEK-ERK (MAPK) pathway and is associated with the abnormal activation and poor prognosis of cancer. To date, several small-molecule inhibitors of RAS, RAF, and MEK have been reported to make rapid advances in cancer therapy; however, acquired resistance still occurs, thereby weakening the therapeutic efficacy of these inhibitors. Recently, selective inhibition of ERK1/2 has been regarded as a potential cancer therapeutic strategy that can not only effectively block the MAPK pathway but also overcome drug resistance caused by upstream mutations in RAS, RAF, and MEK. Herein, we summarize the oncogenic roles, key signaling network, and the single- and dual-target inhibitors of ERK1/2 in preclinical and clinical trials. Together, these inspiring findings shed new light on the discovery of more small-molecule inhibitors of ERK1/2 as candidate drugs to improve cancer therapeutics.
Collapse
Affiliation(s)
- Leilei Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Siwei Chen
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Gu He
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Chen
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Liu
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
Xie S, Wan X, Chen S, Hu Y, Liu X. p21-activated kinase 2 binds to transcription factor SOX2 and up-regulates DEK to promote the progression of lung squamous cell carcinoma. J Transl Med 2022; 102:1109-1120. [PMID: 35821094 DOI: 10.1038/s41374-022-00808-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 11/09/2022] Open
Abstract
Lung squamous cell carcinoma (LSCC) is a prevalent and progressive subtype of lung cancer. This study aimed to substantiate the regulatory effect of the PAK2/SOX2/DEK axis on the LSCC development. LSCC tissues (n = 83) and adjacent normal tissues were collected and SOX2 expression was determined by qRT-PCR and Western blotting. Correlation between SOX2 expression and the prognosis of LSCC patients was then explored utilizing Kaplan-Meier analysis. Co-immunoprecipitation and glutathione-S-transferase pull-down assays were conducted to validate the binding of SOX2 to DEK. Gain- and loss- of function assays were then performed on LSCC cells, with CCK-8 and Transwell assays applied to detect the malignant behaviors of cells. A mouse xenograft model of LSCC was further established for in vivo validation. The expression levels of SOX2, PAK2 and DEK were up-regulated in LSCC tissues and cells. SOX2 overexpression was correlated with poor prognosis of LSCC patients. Knockdown of SOX2 weakened the viability and the migratory and invasive potential of LSCC cells. Further, PAK2 directly interacted with SOX2. PAK2 overexpression accelerated the malignant phenotypes of LSCC cells through interplay with SOX2. Moreover, SOX2 activated the expression of DEK, and silencing DEK attenuated the malignant behaviors of LSCC cells. In conclusion, PAK2 could bind to the transcription factor SOX2 and thus activate the expression of DEK, thereby driving the malignant phenotypes of LSCC cells both in vivo and in vitro.
Collapse
Affiliation(s)
- Shiguang Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 330006, Nanchang, P.R. China
| | - Xuan Wan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 330006, Nanchang, P.R. China
| | - Shuyun Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 330006, Nanchang, P.R. China
| | - Yan Hu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, 330006, Nanchang, P.R. China.
| | - Xiaoming Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, 330006, Nanchang, P.R. China.
| |
Collapse
|
8
|
Barbirou M, Miller A, Manjunath Y, Ramirez AB, Ericson NG, Staveley-O’Carroll KF, Mitchem JB, Warren WC, Chaudhuri AA, Huang Y, Li G, Tonellato PJ, Kaifi JT. Single Circulating-Tumor-Cell-Targeted Sequencing to Identify Somatic Variants in Liquid Biopsies in Non-Small-Cell Lung Cancer Patients. Curr Issues Mol Biol 2022; 44:750-763. [PMID: 35723337 PMCID: PMC8928994 DOI: 10.3390/cimb44020052] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/30/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) accounts for most cancer-related deaths worldwide. Liquid biopsy by a blood draw to detect circulating tumor cells (CTCs) is a tool for molecular profiling of cancer using single-cell and next-generation sequencing (NGS) technologies. The aim of the study was to identify somatic variants in single CTCs isolated from NSCLC patients by targeted NGS. Thirty-one subjects (20 NSCLC patients, 11 smokers without cancer) were enrolled for blood draws (7.5 mL). CTCs were identified by immunofluorescence, individually retrieved, and DNA-extracted. Targeted NGS was performed to detect somatic variants (single-nucleotide variants (SNVs) and insertions/deletions (Indels)) across 65 oncogenes and tumor suppressor genes. Cancer-associated variants were classified using OncoKB database. NSCLC patients had significantly higher CTC counts than control smokers (p = 0.0132; Mann–Whitney test). Analyzing 23 CTCs and 13 white blood cells across seven patients revealed a total of 644 somatic variants that occurred in all CTCs within the same subject, ranging from 1 to 137 per patient. The highest number of variants detected in ≥1 CTC within a patient was 441. A total of 18/65 (27.7%) genes were highly mutated. Mutations with oncogenic impact were identified in functional domains of seven oncogenes/tumor suppressor genes (NF1, PTCH1, TP53, SMARCB1, SMAD4, KRAS, and ERBB2). Single CTC-targeted NGS detects heterogeneous and shared mutational signatures within and between NSCLC patients. CTC single-cell genomics have potential for integration in NSCLC precision oncology.
Collapse
Affiliation(s)
- Mouadh Barbirou
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
| | - Amanda Miller
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
| | - Yariswamy Manjunath
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
| | | | | | - Kevin F. Staveley-O’Carroll
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
| | - Jonathan B. Mitchem
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
| | - Wesley C. Warren
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Department of Animal Sciences and Surgery, Informatics and Data Sciences Institute, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Aadel A. Chaudhuri
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yi Huang
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Guangfu Li
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
| | - Peter J. Tonellato
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
| | - Jussuf T. Kaifi
- Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO 65212, USA; (M.B.); (A.M.); (P.J.T.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Y.M.); (K.F.S.-O.); (J.B.M.); (G.L.)
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65201, USA
- Siteman Cancer Center, St. Louis, MO 63110, USA; (W.C.W.); (A.A.C.); (Y.H.)
- Correspondence:
| |
Collapse
|
9
|
Mapping lung squamous cell carcinoma pathogenesis through in vitro and in vivo models. Commun Biol 2021; 4:937. [PMID: 34354223 PMCID: PMC8342622 DOI: 10.1038/s42003-021-02470-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is the main cause of cancer death worldwide, with lung squamous cell carcinoma (LUSC) being the second most frequent subtype. Preclinical LUSC models recapitulating human disease pathogenesis are key for the development of early intervention approaches and improved therapies. Here, we review advances and challenges in the generation of LUSC models, from 2D and 3D cultures, to murine models. We discuss how molecular profiling of premalignant lesions and invasive LUSC has contributed to the refinement of in vitro and in vivo models, and in turn, how these systems have increased our understanding of LUSC biology and therapeutic vulnerabilities.
Collapse
|
10
|
NELL2 modulates cell proliferation and apoptosis via ERK pathway in the development of benign prostatic hyperplasia. Clin Sci (Lond) 2021; 135:1591-1608. [PMID: 34195782 DOI: 10.1042/cs20210476] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 01/01/2023]
Abstract
Benign prostatic hyperplasia (BPH) is a quite common illness but its etiology and mechanism remain unclear. Neural epidermal growth factor-like like 2 (NELL2) plays multifunctional roles in neural cell growth and is strongly linked to the urinary tract disease. Current study aims to determine the expression, functional activities and underlying mechanism of NELL2 in BPH. Human prostate cell lines and tissues from normal human and BPH patients were utilized. Immunohistochemical staining, immunofluorescent staining, RT-polymerase chain reaction (PCR) and Western blotting were performed. We further generated cell models with NELL2 silenced or overexpressed. Subsequently, proliferation, cycle, and apoptosis of prostate cells were determined by cell counting kit-8 (CCK-8) assay and flow cytometry analysis. The epithelial-mesenchymal transition (EMT) and fibrosis process were also analyzed. Our study revealed that NELL2 was up-regulated in BPH samples and localized in the stroma and the epithelium compartments of human prostate tissues. NELL2 deficiency induced a mitochondria-dependent cell apoptosis, and inhibited cell proliferation via phosphorylating extracellular signal-regulated kinase 1/2 (ERK1/2) activation. Additionally, suppression of ERK1/2 with U0126 incubation could significantly reverse NELL2 deficiency triggered cell apoptosis. Consistently, overexpression of NELL2 promoted cell proliferation and inhibited cell apoptosis. However, NELL2 interference was observed no effect on EMT and fibrosis process. Our novel data demonstrated that up-regulation of NELL2 in the enlarged prostate could contribute to the development of BPH through enhancing cell proliferation and inhibited a mitochondria-dependent cell apoptosis via the ERK pathway. The NELL2-ERK system might represent an important target to facilitate the development of future therapeutic approaches in BPH.
Collapse
|
11
|
ERK1/2: An Integrator of Signals That Alters Cardiac Homeostasis and Growth. BIOLOGY 2021; 10:biology10040346. [PMID: 33923899 PMCID: PMC8072600 DOI: 10.3390/biology10040346] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/24/2022]
Abstract
Integration of cellular responses to extracellular cues is essential for cell survival and adaptation to stress. Extracellular signal-regulated kinase (ERK) 1 and 2 serve an evolutionarily conserved role for intracellular signal transduction that proved critical for cardiomyocyte homeostasis and cardiac stress responses. Considering the importance of ERK1/2 in the heart, understanding how these kinases operate in both normal and disease states is critical. Here, we review the complexity of upstream and downstream signals that govern ERK1/2-dependent regulation of cardiac structure and function. Particular emphasis is given to cardiomyocyte hypertrophy as an outcome of ERK1/2 activation regulation in the heart.
Collapse
|
12
|
Wang Y, Xue Q, Zheng Q, Jin Y, Shen X, Yang M, Zhou X, Li Y. SMAD4 mutation correlates with poor prognosis in non-small cell lung cancer. J Transl Med 2021; 101:463-476. [PMID: 33303972 DOI: 10.1038/s41374-020-00517-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/07/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
SMAD4 is an intracellular signaling mediator of the TGF-β pathway. Its mutation was commonly observed in gastrointestinal cancers, such as pancreatic cancer. The loss of SMAD4 on immunohistochemical staining is often used to suggest a pancreaticobiliary differentiation in evaluating a metastatic adenocarcinoma with unknown origin. However, the function and molecular mechanism of SMAD4 in non-small cell lung cancer (NSCLC) development are largely unknown. Thus, we studied the correlation between SMAD4 mutations and clinico-molecular features in the patients with NSCLC. We reported the frequencies and prognostic values of SMAD4 mutations in a Chinese NSCLC cohort using next-generation sequencing. The NSCLC cases from several public databases, including The Cancer Genome Atlas and others, were also used in this study to elucidate SMAD4-related molecular partners and mechanisms. Integrated bioinformatics analyses were conducted, such as analysis of Gene Ontology enrichment analysis, gene set enrichment analysis (GSEA), and survival analysis. Immunohistochemistry showed that the tissues harboring SMAD4 mutations tended to show SMAD4 deficiency or loss, while SMAD4 expression was significantly reduced at all stages of NSCLC cases. We found that reduced SMAD4 expression was more frequent in the patients with poor disease-free survival and resistance to platinum-based chemotherapy. SMAD4 mutation was an independent risk factor for the survival of NSCLC patients. The expression of SMAD4 was associated with that of SMAD2. The GSEA showed that SMAD4 might promote NSCLC progression by regulating proliferation, adhesion, and immune response. In conclusion, these data suggest that SMAD4 mutation or loss as well as reduced expression can be used to identify the NSCLC patients with poor survival and resistance to platinum-based chemotherapy. SMAD4 may be a predictive marker or therapeutic target in NSCLC. The source code and user's guide are freely available at Github: https://github.com/wangyue77-ab/smad4 .
Collapse
Affiliation(s)
- Yue Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qianqian Xue
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Zheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Jin
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuxia Shen
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mu Yang
- Department of Pathology, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200032, China.
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Guo X, Li M, Wang X, Pan Y, Li J. Correlation between loss of Smad4 and clinical parameters of non-small cell lung cancer: an observational cohort study. BMC Pulm Med 2021; 21:111. [PMID: 33794845 PMCID: PMC8017835 DOI: 10.1186/s12890-021-01480-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Background SMAD4 has been found to be inactivated to varying degrees in many types of cancer; the purpose of this study was to investigate the correlation between SMAD4 expression in non-small cell lung cancer (NSCLC) and clinical pathological parameters. Methods The serum concentration of SMAD4 was measured by enzyme-linked immunosorbent assay and its histological expression was quantified by immunohistochemistry. Results The serum concentration of Smad4 in patients with NSCLC was lower than that in benign lung disease patients and healthy individuals (P < 0.001) and its concentration was related to the histological classification, pathological differentiation, lymphatic metastasis and clinical stage of NSCLC. The sensitivity and specificity of serum Smad4 were 91.56% and 61.56% for screening NSCLC from healthy individuals and 84.55% and 60.36% for screening NSCLC from patients with benign lung disease. Logistic regression analysis showed that the degree of cell differentiation (P < 0.001), lymph node metastasis (P < 0.001) and clinical stage of NSCLC (P = 0.007) affected the expression of Smad4, and had a strong correlation with the expression of Smad4. The expression of Smad4 in NSCLC tissues was lower than that in normal lung tissues (P = 0.009) and its expression was related to the degree of tissue differentiation, lymph node metastasis and clinical stage (P < 0.05). Conclusions The downregulation or deletion of Smad4 is related to the malignant biological behavior of NSCLC and serum Smad4 could be considered as a potential molecular indicator for diagnosis and evaluation of NSCLC.
Collapse
Affiliation(s)
- Xiangjun Guo
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Mengmeng Li
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Xin Wang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Yun Pan
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China
| | - Jiashu Li
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu, China.
| |
Collapse
|
14
|
Dong M, Xu T, Li H, Li X. LINC00052 promotes breast cancer cell progression and metastasis by sponging miR-145-5p to modulate TGFBR2 expression. Oncol Lett 2021; 21:368. [PMID: 33777194 PMCID: PMC7988718 DOI: 10.3892/ol.2021.12629] [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] [Received: 04/07/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) may participate in biological regulatory mechanisms of tumors. The aim of the present study was to uncover the molecular mechanism of the lncRNA LINC00052 in the tumorigenesis of breast cancer (BC). LINC00052 expression in BC tissues and cell lines was detected by reverse transcription-quantitative PCR analysis. The Cell Counting Kit-8, proliferation, Transwell and wound healing assays were employed to confirm the effect of LINC00052 on cell proliferation, migration and invasion. The cell localization of LINC00052 was estimated by cytoplasmic nuclear separation assay. Finally, the potential regulatory mechanism of LINC00052 in BC was detected by western blot analysis. The expression levels of LINC00052 were found to be significantly higher in BC tissues compared with those in the adjacent normal tissues. Downregulation of LINC00052 expression in vitro significantly suppressed the proliferation, migration and invasion of BC cells. LINC00052 was mainly expressed in the cytoplasm and was considered to bind with microRNA (miR)-145-5p based on various databases. Notably, the high expression levels of LINC00052 led to the low expression levels of miR-145-5p and high expression levels of TGF-β receptor II (TGFBR2). In conclusion, the findings of the present study demonstrated that LINC00052 may sponge miR-145-5p to upregulate TGFBR2 expression in order to promote the proliferation and metastasis of BC cells. Therefore, LINC00052 may be an effective potential target for the diagnosis and treatment of BC.
Collapse
Affiliation(s)
- Menglu Dong
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Tao Xu
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hanning Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
15
|
Teicher BA. TGFβ-Directed Therapeutics: 2020. Pharmacol Ther 2021; 217:107666. [PMID: 32835827 PMCID: PMC7770020 DOI: 10.1016/j.pharmthera.2020.107666] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
The transforming growth factor-beta (TGFβ) pathway is essential during embryo development and in maintaining normal homeostasis. During malignancy, the TGFβ pathway is co-opted by the tumor to increase fibrotic stroma, to promote epithelial to mesenchymal transition increasing metastasis and producing an immune-suppressed microenvironment which protects the tumor from recognition by the immune system. Compelling preclinical data demonstrate the therapeutic potential of blocking TGFβ function in cancer. However, the TGFβ pathway cannot be described as a driver of malignant disease. Two small molecule kinase inhibitors which block the serine-threonine kinase activity of TGFβRI on TGFβRII, a pan-TGFβ neutralizing antibody, a TGFβ trap, a TGFβ antisense agent, an antibody which stabilizes the latent complex of TGFβ and a fusion protein which neutralizes TGFβ and binds PD-L1 are in clinical development. The challenge is how to most effectively incorporate blocking TGFβ activity alone and in combination with other therapeutics to improve treatment outcome.
Collapse
Affiliation(s)
- Beverly A Teicher
- Developmental Therapeutics Program, DCTD, National Cancer Institute, RM 4-W602, MSC 9735, 9609 Medical Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Dwyer-Nield LD, McArthur DG, Tennis MA, Merrick DT, Keith RL. An Improved Murine Premalignant Squamous Cell Model: Tobacco Smoke Exposure Augments NTCU-Induced Murine Airway Dysplasia. Cancer Prev Res (Phila) 2020; 14:307-312. [PMID: 33115782 DOI: 10.1158/1940-6207.capr-20-0332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/25/2020] [Accepted: 10/22/2020] [Indexed: 12/31/2022]
Abstract
Tobacco smoke-induced squamous cell lung cancer (SCC) develops from endobronchial dysplastic lesions that progress to invasive disease. A reproducible murine model recapitulating histologic progression observed in current and former smokers will advance testing of new preventive and therapeutic strategies. Previous studies show that prolonged topical application of N-nitroso-tris-chloroethylurea (NTCU) generates a range of airway lesions in sensitive mice similar to those induced by chronic tobacco smoke exposure in humans. To improve the current NTCU model and better align it with human disease, NTCU was applied to mice twice weekly for 4-5 weeks followed by a recovery period before cigarette smoke (CS) or ambient air (control) exposure for an additional 3-6 weeks. Despite the short time course, the addition of CS led to significantly more premalignant lesions (PML; 2.6 vs. 0.5; P < 0.02) and resulted in fewer alveolar macrophages (52,000 macrophages/mL BALF vs. 68,000; P < 0.05) compared with control mice. This improved NTCU + CS model is the first murine SCC model to incorporate tobacco smoke and is more amenable to preclinical studies because of the increased number of PML, decreased number of mice required, and reduced time needed for PML development.
Collapse
Affiliation(s)
- Lori D Dwyer-Nield
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Debbie G McArthur
- Research Division, Rocky Mountain Regional Veterans Administration Medical Center, Aurora, Colorado
| | - Meredith A Tennis
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Daniel T Merrick
- Division of Pathology, School of Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado
| | - Robert L Keith
- Research Division, Rocky Mountain Regional Veterans Administration Medical Center, Aurora, Colorado. .,Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Center, Aurora, Colorado
| |
Collapse
|
17
|
Yang Y, Fan X, Ren Y, Wu K, Tian X, Wen F, Liu D, Fan Y, Zhao S. SOX2-Upregulated microRNA-30e Promotes the Progression of Esophageal Cancer via Regulation of the USP4/SMAD4/CK2 Axis. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 23:200-214. [PMID: 33376627 PMCID: PMC7750169 DOI: 10.1016/j.omtn.2020.10.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Esophageal cancer (EC) is a highly aggressive disease, and its progression involves a complex gene regulation network. Transcription factor SOX2 is amplified in various cancers including EC. A pathway involving SOX2 regulation of microRNAs (miRNAs) and their target genes has been previously revealed. This study aims to delineate the ability of SOX2 to influence the EC progression, with the involvement of miR-30e/USP4/SMAD4/CK2 axis. SOX2 expression was first examined in the clinical tissue samples from 30 EC patients. Effects of SOX2 on proliferation, migration, and invasion alongside tumorigenicity of transfected cells were examined by means of gain- and loss-of-function experiments. EC tissues and cells exhibited high expression of SOX2, miR-30e, and CK2 and poor expression of USP4 and SMAD4. Mechanistically, SOX2 was positively correlated with miR-30e and upregulated the expression of miR-30e. miR-30e specifically targeted USP4, which induced deubiquitination of SMAD4 and promoted its expression. Meanwhile, SMAD4 was enriched in the CK2 promoter region and thus inhibited its expression. SOX2 stimulated EC cell proliferative, invasive, and migratory capacities in vitro and tumor growth in vivo by regulating the miR-30e/USP4/SMAD4/CK2 axis. Collectively, our work reveals a novel SOX2-mediated regulatory network in EC that may be a viable target for EC treatment.
Collapse
Affiliation(s)
- Yang Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China
| | - Xin Fan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China
| | - Yukai Ren
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China
| | - Kai Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China
| | - Xiangyu Tian
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China
| | - Fengbiao Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China
| | - Donglei Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China
| | - Yuxia Fan
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China
- Corresponding author Yuxia Fan, Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China.
| | - Song Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China
- Corresponding author Song Zhao, Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, P.R. China.
| |
Collapse
|
18
|
Liu Y, Jia W, Li J, Zhu H, Yu J. Identification of Survival-Associated Alternative Splicing Signatures in Lung Squamous Cell Carcinoma. Front Oncol 2020; 10:587343. [PMID: 33117720 PMCID: PMC7561379 DOI: 10.3389/fonc.2020.587343] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 08/28/2020] [Indexed: 02/05/2023] Open
Abstract
Purpose: Alternative splicing (AS) is a post-transcriptional process that plays a significant role in enhancing the diversity of transcription and protein. Accumulating evidences have demonstrated that dysregulation of AS is associated with oncogenic processes. However, AS signature specifically in lung squamous cell carcinoma (LUSC) remains unknown. This study aimed to evaluate the prognostic values of AS events in LUSC patients. Methods: The RNA-seq data, AS events data and corresponding clinical information were obtained from The Cancer Genome Atlas (TCGA) database. Univariate Cox regression analysis was performed to identify survival-related AS events and survival-related parent genes were subjected to Gene Ontology enrichment analysis and gene network analysis. The least absolute shrinkage and selection operator (LASSO) method and multivariate Cox regression analysis were used to construct prognostic prediction models, and their predictive values were assessed by Kaplan-Meier analysis and receiver operating characteristic (ROC) curves. Then a nomogram was established to predict the survival of LUSC patients. And the interaction network of splicing factors (SFs) and survival-related AS events was constructed by Spearman correlation analysis and visualized by Cytoscape. Results: Totally, 467 LUSC patients were included in this study and 1,991 survival-related AS events within 1,433 genes were identified. SMAD4, FOS, POLR2L, and RNPS1 were the hub genes in the gene interaction network. Eight prognostic prediction models (seven types of AS and all AS) were constructed and all exhibited high efficiency in distinguishing good or poor survival of LUSC patients. The final integrated prediction model including all types of AS events exhibited the best prognostic power with the maximum AUC values of 0.778, 0.816, 0.814 in 1, 3, 5 years ROC curves, respectively. Meanwhile, the nomogram performed well in predicting the 1-, 3-, and 5-year survival of LUSC patients. In addition, the SF-AS regulatory network uncovered a significant correlation between SFs and survival-related AS events. Conclusion: This is the first comprehensive study to analyze the role of AS events in LUSC specifically, which improves our understanding of the prognostic value of survival-related AS events for LUSC. And these survival-related AS events might serve as novel prognostic biomarkers and drug therapeutic targets for LUSC.
Collapse
Affiliation(s)
- Yang Liu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wenxiao Jia
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ji Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
19
|
Yuan J, Yi K, Yang L. TGFBR2 Regulates Hedgehog Pathway and Cervical Cancer Cell Proliferation and Migration by Mediating SMAD4. J Proteome Res 2020; 19:3377-3385. [PMID: 32628850 DOI: 10.1021/acs.jproteome.0c00239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Jialing Yuan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No.
20, Section 3, Renmin Nanlu Road, Chengdu, Sichuan 610041, China
| | - Ke Yi
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No.
20, Section 3, Renmin Nanlu Road, Chengdu, Sichuan 610041, China
| | - Lingyun Yang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No.
20, Section 3, Renmin Nanlu Road, Chengdu, Sichuan 610041, China
| |
Collapse
|
20
|
Li F, Li F, Chen W. Propofol Inhibits Cell Proliferation, Migration, and Invasion via mir-410-3p/Transforming Growth Factor-β Receptor Type 2 (TGFBR2) Axis in Glioma. Med Sci Monit 2020; 26:e919523. [PMID: 31960827 PMCID: PMC6993559 DOI: 10.12659/msm.919523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Propofol is a common intravenous anesthetic used to induce and maintain anesthesia. Numerous studies have reported that propofol plays an anti-tumor role in diverse human cancers, including glioma. In this research, we explored the roles of propofol and its related molecular mechanisms in glioma. MATERIAL AND METHODS U251 and A172 cells were exposed to different doses of propofol for 24 h. Cell proliferation, migration, and invasion in glioma were evaluated using MTT assay and Transwell assay, respectively. The levels of microRNA-410-3p (miR-410-3p) and transforming growth factor-ß receptor type 2 (TGFBR2) were detected by quantitative real-time polymerase chain reaction (qRT-PCR) assay and Western blot assay, respectively. The association between miR-410-3p and TGFBR2 was predicted by TargetScan and confirmed by dual-luciferase reporter assay. RESULTS Propofol inhibited the proliferation, migration, and invasion of glioma cells in a concentration-dependent way. miR-410-3p was induced and TGFBR2 was inhibited by different concentrations of propofol treatment. Moreover, TGFBR2 was confirmed to be a target gene of miR-410-3p and TGFBR2 was inversely modulated by miR-410-3p in glioma cells. Depletion of miR-410-3p reversed the inhibition of propofol treatment on U251 and A172 cell growth and metastasis, but the effects were further abolished by knocking down the expression of TGFBR2. CONCLUSIONS Propofol can suppress cell growth and metastasis by regulating the miR-410-3p/TGFBR2 axis in glioma.
Collapse
Affiliation(s)
- Fengli Li
- Department of Anesthesiology, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| | - Fengliang Li
- Department of Psychiatry, Third Hospital of Weifang, Weifang, Shandong, China (mainland)
| | - Wei Chen
- Department of Neurosurgery, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| |
Collapse
|
21
|
Chao J, Jin L, Zhang X, Ding D, Wu S, Ma L, Zhu B, Shan S, Yun X, Gao P, Li J, Zhu C, Qin X. Insight into the effects of microRNA-23a-3p on pancreatic cancer and its underlying molecular mechanism. Oncol Lett 2019; 19:187-194. [PMID: 31897129 PMCID: PMC6924110 DOI: 10.3892/ol.2019.11117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 09/24/2019] [Indexed: 12/14/2022] Open
Abstract
Previous studies have demonstrated that microRNA (miR)-23a-3p plays a role as an oncogene that is involved in several different types of carcinoma. However, few studies investigated the association between miR-23a-3p and pancreatic cancer (PC). The aim of the present study was to elucidate the biological functions of miR-23a-3p in PC and to investigate its underlying molecular mechanisms. The expression of miR-23a-3p in PC and adjacent normal tissues was investigated using microarrays. In order to validate the outcomes of the microarray results, reverse transcription-quantitative (RT-q)PCR was used to determine the expression levels of miR-23a-3p in PC tissues and cell lines. Furthermore, functional analyses were conducted following miR-23a-3p inhibition and overexpression, in order to assess the proliferation, invasion and migration of PC cells. Bioinformatics analysis indicated transforming growth factor-β receptor type II (TGFBR2) as a potential direct target of miR-23a-3p. Western blotting was performed in order to determine the protein expression of TGFBR2 in PC cell lines. The findings from the microarray demonstrated upregulation of miR-23a-3p in PC compared with normal tissues. RT-qPCR revealed significantly higher levels of miR-23a-3p expression in PC compared with normal control tissues or cells. Furthermore, miR-23a-3p was demonstrated to promote the proliferation, invasion and migration of PC cells, which was suppressed by the inhibition of miR-23a-3p. In addition, the miR-23a-3p expression level was negatively associated with TGFBR2 expression. Overall, the present study demonstrated the tumor-promoting effects of miR-23a-3p in PC cells. Furthermore, miR-23a-3p is a potential oncogenic regulator of PC, by targeting TGFBR2, and a biomarker or target for molecular therapy.
Collapse
Affiliation(s)
- Jiadeng Chao
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Lei Jin
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Xudong Zhang
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Dong Ding
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Siyuan Wu
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Le Ma
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Bei Zhu
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Shiting Shan
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Xiao Yun
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Peng Gao
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Jun Li
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Chunfu Zhu
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Xihu Qin
- Department of Pancreatopathy, Pancreas Center of Changzhou Clinical College, Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China.,Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|