1
|
Jin F, Lin Y, Yuan W, Wu S, Yang M, Ding S, Liu J, Chen Y. Recent advances in c-Met-based dual inhibitors in the treatment of cancers. Eur J Med Chem 2024; 272:116477. [PMID: 38733884 DOI: 10.1016/j.ejmech.2024.116477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
The cellular-mesenchymal epithelial transition factor (c-Met) is a receptor tyrosine kinase (RTK) located on the 7q31 locus encoding the Met proto-oncogene and plays a critical role in regulating cell proliferation, metastasis, differentiation, and apoptosis through various signaling pathways. However, its aberrant activation and overexpression have been implicated in many human cancers. Therefore, c-Met is a promising target for cancer treatment. However, the anticancer effect of selective single-targeted drugs is limited due to the complexity of the signaling system and the involvement of different proteins and enzymes. After inhibiting one pathway, signal molecules can be transmitted through other pathways, resulting in poor efficacy of single-targeted drug therapy. Dual inhibitors that simultaneously block c-Met and another factor can significantly improve efficacy and overcome some of the shortcomings of single-target inhibitors, including drug resistance. In this review, We introduced c-Met kinase and the synergism between c-Met and other anti-tumor targets, then dual-target inhibitors based on c-Met for the treatment of cancers were summarized and their design concepts and structure-activity relationships (SARs) were discussed elaborately, providing a valuable insight for the further development of novel c-Met-based dual inhibitors.
Collapse
Affiliation(s)
- Fanqi Jin
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China
| | - Yihan Lin
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China
| | - Weidong Yuan
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China
| | - Shuang Wu
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China
| | - Min Yang
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China
| | - Shi Ding
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China; API Engineering Technology Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China; Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China
| | - Ju Liu
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China; API Engineering Technology Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China; Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China.
| | - Ye Chen
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China; API Engineering Technology Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China; Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China.
| |
Collapse
|
2
|
Zhao HF, Liu YS, Wang J, Wu CP, Zhou XM, Cai LR, Liu J, Liu XJ, Xu YW, Li WP, Huang GD. Nuclear transport of phosphorylated LanCL2 promotes invadopodia formation and tumor progression of glioblastoma by activating STAT3/Cortactin signaling. J Adv Res 2024:S2090-1232(24)00107-3. [PMID: 38492734 DOI: 10.1016/j.jare.2024.03.007] [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: 01/23/2024] [Revised: 02/28/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024] Open
Abstract
INTRODUCTION Our previous study showed that the abscisic acid receptor lanthionine synthetase C-like 2 (LanCL2) is a significant prognostic factor for overall survival in young glioblastoma patients. However, the role of LanCL2 in glioblastoma remains unclear yet. OBJECTIVES This study aims to investigate the role of LanCL2 in regulating in-vitro cell invasion and in-vivo tumor progression of glioblastoma and its underlying mechanism. METHODS Tyrosine 198 or 295 residue of LanCL2 was mutated using site-directed mutagenesis to block its phosphorylation. The role of LanCL2 in glioblastoma was investigated using transwell or 3D invasion assay, matrix degradation assay and intracranial xenograft model. RESULTS This study showed that nuclear transport of LanCL2 was enhanced by overexpression of LanCL2 or its ligand abscisic acid in glioblastoma cells. Knockdown of LanCL2 suppressed migration, invasion and invadopodia formation of glioblastoma cells, whereas overexpression of wild-type LanCL2 enhanced them. Blocking of Tyr295 residue phosphorylation of LanCL2 impeded its nuclear transport, retarded glioblastoma cell motility and invadopodia formation, and deceased the phosphorylation of Cortactin and STAT3. c-Met was identified as the upstream tyrosine kinase of Tyr295 residue of LanCL2, and inhibition of c-Met markedly suppressed the nuclear transport of LanCL2. Moreover, overexpression of wild-type LanCL2 significantly promoted orthotopic tumor growth of glioblastoma in vivo and led to poor survival of mice with median survival time of 33.5 days, whereas Tyr295 mutation rescued it with median survival time of 49 days. CONCLUSION Our findings suggested that Tyr295 phosphorylation is crucial to the activation and nuclear transport of LanCL2, as well as invadopodia formation and tumor progression of glioblastoma, providing the evidence of a novel signaling axis c-Met/LanCL2/STAT3/Cortactin and the first observation of the importance of Tyr295 phosphorylation to LanCL2.
Collapse
Affiliation(s)
- Hua-Fu Zhao
- Department of Neurosurgery, Institute of Translational Medicine, Shenzhen University First Affiliated Hospital, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| | - Yun-Sheng Liu
- Department of Neurosurgery, Institute of Translational Medicine, Shenzhen University First Affiliated Hospital, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Jing Wang
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chang-Peng Wu
- Department of Neurosurgery, Shenzhen Longhua New District People's Hospital, Shenzhen 518109, China
| | - Xiu-Ming Zhou
- Epilepsy Center, Guangdong 999 Brain Hospital, Guangzhou 510510, China
| | - Lin-Rong Cai
- Department of Neurosurgery, Institute of Translational Medicine, Shenzhen University First Affiliated Hospital, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Jing Liu
- Department of Pathology, Shenzhen University First Affiliated Hospital, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Xiao-Jia Liu
- Department of Neurosurgery, Institute of Translational Medicine, Shenzhen University First Affiliated Hospital, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Yan-Wen Xu
- Department of Neurosurgery, Institute of Translational Medicine, Shenzhen University First Affiliated Hospital, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Wei-Ping Li
- Department of Neurosurgery, Institute of Translational Medicine, Shenzhen University First Affiliated Hospital, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Guo-Dong Huang
- Department of Neurosurgery, Institute of Translational Medicine, Shenzhen University First Affiliated Hospital, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| |
Collapse
|
3
|
An B, Nie W, Hu J, Fan Y, Nie H, Wang M, Zhao Y, Yao H, Ren Y, Zhang C, Wei M, Li W, Liu J, Yang C, Zhang Y, Li X, Tian G. A novel c-Met/TRK inhibitor 1D228 efficiently inhibits tumor growth by targeting angiogenesis and tumor cell proliferation. Cell Death Dis 2023; 14:728. [PMID: 37945598 PMCID: PMC10636171 DOI: 10.1038/s41419-023-06246-5] [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: 04/19/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
Multiple tumors are synergistically promoted by c-Met and TRK, and blocking their cross-signalling pathway may give better effects. In this study, we developed a tyrosine kinase inhibitor 1D228, which exhibited excellent anti-tumor activity by targeting c-Met and TRK. Models in vitro, 1D228 showed a significant better inhibition on cancer cell proliferation and migration than the positive drug Tepotinib. Models in vivo, 1D228 showed robust anti-tumor effect on gastric and liver tumor growth with 94.8% and 93.4% of the TGI, respectively, comparing 67.61% and 63.9% of Tepotinib. Importantly, compared with the combination of Larotrectinib and Tepotinib, 1D228 monotherapy in MKN45 xenograft tumor models showed stronger antitumor activity and lower toxicity. Mechanistic studies showed that 1D228 can largely inhibit the phosphorylation of TRKB and c-Met. Interestingly, both kinases, TRKs and c-Met, have been found to be co-expressed at high levels in patients with gastric cancer through IHC. Furthermore, bioinformatics analysis has revealed that both genes are abnormally co-expressed in multiple types of cancer. Cell cycle analysis found that 1D228 induced G0/G1 arrest by inhibiting cyclin D1. Additionally, vascular endothelial cells also showed a pronounced response to 1D228 due to its expression of TRKB and c-Met. 1D228 suppressed the migration and tube formation of endothelial cells, which are the key functions of tumor angiogenesis. Taken together, compound 1D228 may be a promising candidate for the next generation of c-Met and TRK inhibitors for cancer treatment, and offers a novel potential treatment strategy for cancer patients with abnormal expressions of c-Met or NTRK, or simultaneous of them.
Collapse
Affiliation(s)
- Baijiao An
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Wenyan Nie
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Jinhui Hu
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Yangyang Fan
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Haoran Nie
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Mengxuan Wang
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Yaxuan Zhao
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Han Yao
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Yuanyuan Ren
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Chuanchuan Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Mengna Wei
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Wei Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Jiadai Liu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China
| | - Chunhua Yang
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China
| | - Yin Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China.
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Yantai, Shandong, 264003, PR China.
| | - Xingshu Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, PR China.
| | - Geng Tian
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, PR China.
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Yantai, Shandong, 264003, PR China.
| |
Collapse
|
4
|
Gallo S, Spilinga M, Albano R, Ferrauto G, Di Gregorio E, Casanova E, Balmativola D, Bonzano A, Boccaccio C, Sapino A, Comoglio PM, Crepaldi T. Activation of the MET receptor attenuates doxorubicin-induced cardiotoxicity in vivo and in vitro. Br J Pharmacol 2020; 177:3107-3122. [PMID: 32133617 PMCID: PMC7280013 DOI: 10.1111/bph.15039] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Background and Purpose Doxorubicin anti‐cancer therapy is associated with cardiotoxicity, resulting from DNA damage response (DDR). Hepatocyte growth factor (HGF) protects cardiomyocytes from injury, but its effective use is compromised by low biodistribution. In this study, we have investigated whether the activation of the HGF receptor—encoded by the Met gene—by an agonist monoclonal antibody (mAb) could protect against doxorubicin‐induced cardiotoxicity. Experimental Approach The mAb (5 mg·kg−1) was injected in vivo into C57BL/6J mice, before doxorubicin (three doses of 7 mg·kg−1). Cardiac functions were evaluated through MRI after treatment termination. Heart histological staining and mRNA levels of genes associated with heart failure (Acta1 and Nppa), inflammation (IL‐6), and fibrosis (Ctgf, Col1a2, Timp1, and Mmp9) were assessed. MAb (100 nM) was administered in vitro to H9c2 cardiomyoblasts before addition of doxorubicin (25 μM). DDR and apoptosis markers were evaluated by quantitative western blotting, flow cytometry, and immunofluorescence. Stattic was used for pharmacological inactivation of STAT3. Key Results In vivo, administration of the mAb alleviated doxorubicin‐induced cardiac dysfunction and fibrosis. In vitro, mAb mimicked the response to HGF by (a) inhibiting histone H2AX phosphorylation at S139, (b) quenching the expression of the DNA repair enzyme PARP1, and (c) reducing the proteolytic activation of caspase 3. The MET‐driven cardioprotection involved, at least in vitro, the phosphorylation of STAT3. Conclusion and Implications The MET agonist mAb provides a new tool for cardioprotection against anthracycline cardiotoxicity.
Collapse
Affiliation(s)
- Simona Gallo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Martina Spilinga
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| | | | - Giuseppe Ferrauto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Enza Di Gregorio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Elena Casanova
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | | | | | - Carla Boccaccio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Tiziana Crepaldi
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| |
Collapse
|
5
|
Liu B, Liu Q, Pan S, Huang Y, Qi Y, Li S, Xiao Y, Jia L. The HOTAIR/miR-214/ST6GAL1 crosstalk modulates colorectal cancer procession through mediating sialylated c-Met via JAK2/STAT3 cascade. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:455. [PMID: 31694696 PMCID: PMC6836492 DOI: 10.1186/s13046-019-1468-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/23/2019] [Indexed: 12/31/2022]
Abstract
Background The regulatory non-coding RNAs, including long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), emerge as pivotal markers during tumor progression. Abnormal sialylated glycoprotein often leads to the malignancy of colorectal cancer (CRC). Methods Differential levels of HOTAIR and ST6GAL1 are analyzed by qRT-PCR. Functionally, CRC cell proliferation, aggressiveness and apoptosis are measured through relevant experiments, including CCK8 assay, colony formation assay, transwell assay, western blot and flow cytometry. Dual-luciferase reporter gene assay and RIP assay confirm the direct interaction between HOTAIR and miR-214. The lung metastasis, liver metatstasis and xenografts nude mice models are established to show the in vivo effect of HOATIR. Results Here, differential levels of HOTAIR and ST6GAL1 are primarily observed in CRC samples and cells. Upregulated HOTAIR and ST6GAL1 are crucial predictors for poor CRC prognosis. Altered level of ST6GAL1 modulates CRC malignancy. Furthermore, ST6GAL1 and HOTAIR are confirmed as the direct targets of miR-214, and ST6GAL1 is regulated by HOTAIR via sponging miR-214. ST6GAL1 induces the elevated metabolic sialylation of c-Met, which is co-mediated by HOTAIR and miR-214. Sialylated c-Met affects the activity of JAK2/STAT3 pathway. The regulatory role of HOTAIR/miR-214/ST6GAL1 axis also impacts CRC procession. In addition, HOTAIR mediates lung metastasis, liver metastasis and tumorigenesis in vivo. ShHOTAIR and AMG-208 are combined to inhibit tumorigenesis for successful drug development. Conclusion The HOTAIR/miR-214/ST6GAL1 axis commands the CRC malignancy by modifying c-Met with sialylation and activating JAK2/STAT3 pathway. Our study presents novel insights into CRC progression and provided prospective therapeutic target for CRC.
Collapse
Affiliation(s)
- Bing Liu
- College of Laboratory Medicine, Dalian Medical University, 9 Lushunnan Road Xiduan, Dalian, 116044, Liaoning Province, China
| | - Qianqian Liu
- College of Laboratory Medicine, Dalian Medical University, 9 Lushunnan Road Xiduan, Dalian, 116044, Liaoning Province, China
| | - Shimeng Pan
- College of Laboratory Medicine, Dalian Medical University, 9 Lushunnan Road Xiduan, Dalian, 116044, Liaoning Province, China
| | - Yiran Huang
- College of Laboratory Medicine, Dalian Medical University, 9 Lushunnan Road Xiduan, Dalian, 116044, Liaoning Province, China
| | - Yu Qi
- College of Laboratory Medicine, Dalian Medical University, 9 Lushunnan Road Xiduan, Dalian, 116044, Liaoning Province, China
| | - Shuangda Li
- College of Laboratory Medicine, Dalian Medical University, 9 Lushunnan Road Xiduan, Dalian, 116044, Liaoning Province, China
| | - Yang Xiao
- College of Laboratory Medicine, Dalian Medical University, 9 Lushunnan Road Xiduan, Dalian, 116044, Liaoning Province, China
| | - Li Jia
- College of Laboratory Medicine, Dalian Medical University, 9 Lushunnan Road Xiduan, Dalian, 116044, Liaoning Province, China.
| |
Collapse
|
6
|
Chen W, Zhang H, Zeng X, Chen L, Fang G, Cai H, Zhong X. Phase‐shifted pentafluorobutane nanoparticles for ultrasound imaging and ultrasound‐mediated hypoxia modulation. J Cell Biochem 2019; 120:16543-16552. [PMID: 31099025 DOI: 10.1002/jcb.28914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 03/18/2019] [Accepted: 04/05/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Wei‐Jian Chen
- Department of Ultrasonography The First Affiliated Hospital of Jinan University Guangzhou China
| | - Hua Zhang
- The First Affiliated Hospital, Biomedical Translation Research Institute and School of Pharmacy Jinan University Guangzhou China
| | - Xue‐Yi Zeng
- Department of Chemistry, College of Chemistry and Materials Science Jinan University Guangzhou China
| | - Long Chen
- Department of Ultrasonography, Xiangyang Central Hospital Affiliated Hospital of Hubei University of Arts and Science Xiangyang China
| | - Gui‐Ting Fang
- Department of Ultrasonography The First Affiliated Hospital of Jinan University Guangzhou China
| | - Huai‐Hong Cai
- Department of Chemistry, College of Chemistry and Materials Science Jinan University Guangzhou China
| | - Xing Zhong
- Department of Ultrasonography The First Affiliated Hospital of Jinan University Guangzhou China
| |
Collapse
|
7
|
Luo T, Zhang SG, Zhu LF, Zhang FX, Li W, Zhao K, Wen XX, Yu M, Zhan YQ, Chen H, Ge CH, Gao HY, Wang L, Yang XM, Li CY. A selective c-Met and Trks inhibitor Indo5 suppresses hepatocellular carcinoma growth. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:130. [PMID: 30885237 PMCID: PMC6421704 DOI: 10.1186/s13046-019-1104-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/11/2019] [Indexed: 11/18/2022]
Abstract
Background Human hepatocellular carcinoma (HCC) lacks effective curative therapy and there is an urgent need to develop a novel molecular-targeted therapy for HCC. Selective tyrosine kinase inhibitors have shown promise in treating cancers including HCC. Tyrosine kinases c-Met and Trks are potential therapeutic targets of HCC and strategies to interrupt c-Met and Trks cross-signaling may result in increased effects on HCC inhibition. Methods The effects of Indo5 on c-Met and Trks activity were determined with in vitro kinase activity assay, cell-based signaling pathway activation, and kinases-driven cell transformation. The in vivo anti-tumor activity was determined with xenograft mice and liver orthotopic mice models. The co-expression of c-Met and TrkB in 180 pairs of HCC and adjacent normal tissues were detected using immunohistochemical staining. Results Indo5, a novel lead compound displayed biochemical potency against both c-Met and Trks with selectivity over 13 human kinases. Indo5 abrogated HGF-induced c-Met signaling activation and BDNF/NGF-induced Trks signal activation, c-Met or TrkB-mediated cell transformation and migration. Furthermore, Indo5 significantly decreased the growth of HCC cells in xenograft mice and improved the survival of mice with liver orthotopic tumors. In addition, co-expression of c-Met and TrkB in HCC patients was a predictor of poor prognosis, and combined inhibition of c-Met and TrkB exerted a synergistic suppressive effect on HCC. Conclusions These findings indicate that Indo5 is associated with marked suppression of c-Met and Trks co-expressing HCC, supporting its clinical development as an antitumor treatment for HCC patients with co-active c-Met and Trks signaling. Electronic supplementary material The online version of this article (10.1186/s13046-019-1104-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Teng Luo
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.,Beijing Institute of Radiation Medicine, 27-Taiping Road, Beijing, 100850, People's Republic of China.,Institute of NBC Defence, Beijing, 102205, China
| | - Shou-Guo Zhang
- Beijing Institute of Radiation Medicine, 27-Taiping Road, Beijing, 100850, People's Republic of China
| | | | - Fei-Xiang Zhang
- Guangdong pharmaceutical university, School of Pharmacy, Guangzhou, 510006, China
| | - Wei Li
- Beijing Institute of Radiation Medicine, 27-Taiping Road, Beijing, 100850, People's Republic of China
| | - Ke Zhao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Xiao-Xue Wen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Miao Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Yi-Qun Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Hui Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Chang-Hui Ge
- Beijing Institute of Radiation Medicine, 27-Taiping Road, Beijing, 100850, People's Republic of China
| | - Hui-Ying Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Lin Wang
- Beijing Institute of Radiation Medicine, 27-Taiping Road, Beijing, 100850, People's Republic of China.
| | - Xiao-Ming Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China. .,School of Chemical Engineering and Technology, Department of pharmaceutical engineering, Tianjin University, Tianjin, 300072, China.
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China. .,An Hui Medical University, Hefei, 230032, China. .,Guangdong pharmaceutical university, School of Pharmacy, Guangzhou, 510006, China.
| |
Collapse
|
8
|
Verma N, Keinan O, Selitrennik M, Karn T, Filipits M, Lev S. PYK2 sustains endosomal-derived receptor signalling and enhances epithelial-to-mesenchymal transition. Nat Commun 2015; 6:6064. [PMID: 25648557 DOI: 10.1038/ncomms7064] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 12/10/2014] [Indexed: 12/11/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a central developmental process implicated in cancer metastasis. Here we show that the tyrosine kinase PYK2 enhances cell migration and invasion and potentiates EMT in human breast carcinoma. EMT inducer, such as EGF, induces rapid phosphorylation of PYK2 and its translocation to early endosomes where it co-localizes with EGFR and sustains its downstream signals. Furthermore, PYK2 enhances EGF-induced STAT3-phosphorylation, while phospho-STAT3 directly binds to PYK2 promoter and regulates PYK2 transcription. STAT3 and PYK2 also enhance c-Met expression, while c-Met augments their phosphorylation, suggesting a positive feedback loop between PYK2-STAT3-c-Met. We propose that PYK2 sustains endosomal-derived receptor signalling and participates in a positive feedback that links cell surface receptor(s) to transcription factor(s) activation, thereby prolonging signalling duration and potentiating EMT. Given the role of EMT in breast cancer metastasis, we also found a significant correlation between PYK2 expression, tumour grade and lymph node metastasis, thus, demonstrating the clinicopathological implication of our findings.
Collapse
Affiliation(s)
- Nandini Verma
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Omer Keinan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Michael Selitrennik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Thomas Karn
- Department of Obstetrics and Gynecology, Goethe University Frankfurt, Theodor-Stern Kai 7, 60590 Frankfurt, Germany
| | - Martin Filipits
- Clinical Division of Oncology, Department of Medicine I, University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Sima Lev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
9
|
Miranda A, López-Cardona AP, Laguna-Barraza R, Calle A, López-Vidriero I, Pintado B, Gutiérrez-Adán A. Transcriptome profiling of liver of non-genetic low birth weight and long term health consequences. BMC Genomics 2014; 15:327. [PMID: 24884990 PMCID: PMC4229907 DOI: 10.1186/1471-2164-15-327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 04/23/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND It is believed that the main factors of low prenatal growth in mammals are genetic and environmental. We used isogenic mice maintained in standard conditions to analyze how natural non-genetic microsomia (low birth weight) is produced in inbred mice and its long term effect on health. To better understand the molecular basis of non-genetic microsomia, we undertook transcriptome profiling of both male and female livers from small and normal size mice at birth. RESULTS Naturally occurring neonatal microsomia was defined as a gender-specific weanling weight under the 10th percentile of the colony. Birth weight variation was similar in inbred and outbred lines. Mice were phenotyped by weight, size, blood pressure, organ size, their response to a glucose challenge, and survival rates. Regardless of diet, adult mice born with microsomia showed a significantly lower body weight and size, and differences in the weight of several organs of microsomic adult mice compared to normal birth weight adults were found. After a high-fat diet, microsomic mice were less prone to obesity, showing a better glucose tolerance and lower blood pressure. Through a transcriptome analysis, we detected a different pattern of mRNA transcription in the liver at birth comparing male vs female and microsomic vs normal mice, noting some modifications in epigenetic regulatory genes in females and modifications in some growth factor genes in males. Finally, using embryo transfer of embryos of different quality and age, we identified a putative preimplantation origin of this non-genetic microsomia. CONCLUSIONS (1) neonatal microsomia is not always a risk factor for adult metabolic syndrome, (2) neonatal non-genetic microsomia displays changes in the expression of important epigenetic genes and changes in liver mRNA transcription profile at birth, exaggerating sexual dimorphism, and (3) random preimplantation phenotypic variability could partially explain body birth weight variation in isogenic lines.
Collapse
Affiliation(s)
- Alberto Miranda
- Dpto, de Reproducción Animal, INIA, Avda Puerta de Hierro no, 12, Local 10, Madrid 28040, Spain.
| | | | | | | | | | | | | |
Collapse
|
10
|
Augsten M, Böttcher A, Spanbroek R, Rubio I, Friedrich K. Graded inhibition of oncogenic Ras-signaling by multivalent Ras-binding domains. Cell Commun Signal 2014; 12:1. [PMID: 24383791 PMCID: PMC3898410 DOI: 10.1186/1478-811x-12-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 12/26/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Ras is a membrane-associated small G-protein that funnels growth and differentiation signals into downstream signal transduction pathways by cycling between an inactive, GDP-bound and an active, GTP-bound state. Aberrant Ras activity as a result of oncogenic mutations causes de novo cell transformation and promotes tumor growth and progression. RESULTS Here, we describe a novel strategy to block deregulated Ras activity by means of oligomerized cognate protein modules derived from the Ras-binding domain of c-Raf (RBD), which we named MSOR for multivalent scavengers of oncogenic Ras. The introduction of well-characterized mutations into RBD was used to adjust the affinity and hence the blocking potency of MSOR towards activated Ras. MSOR inhibited several oncogenic Ras-stimulated processes including downstream activation of Erk1/2, induction of matrix-degrading enzymes, cell motility and invasiveness in a graded fashion depending on the oligomerization grade and the nature of the individual RBD-modules. The amenability to accurate experimental regulation was further improved by engineering an inducible MSOR-expression system to render the reversal of oncogenic Ras effects controllable. CONCLUSION MSOR represent a new tool for the experimental and possibly therapeutic selective blockade of oncogenic Ras signals.
Collapse
Affiliation(s)
- Martin Augsten
- Department of Oncology-Pathology, Karolinska Institutet, 171 76, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
11
|
Shi MD, Liao YC, Shih YW, Tsai LY. Nobiletin attenuates metastasis via both ERK and PI3K/Akt pathways in HGF-treated liver cancer HepG2 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:743-752. [PMID: 23537747 DOI: 10.1016/j.phymed.2013.02.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 01/03/2013] [Accepted: 02/19/2013] [Indexed: 06/02/2023]
Abstract
Hepatocyte growth factor (HGF), and its receptor, c-Met activation has recently been shown to play important roles in cancer invasion and metastasis in a wide variety of tumor cells. We use HGF as an invasive inducer of human HepG2 cells to investigate the effect of four flavones including apigenin, tricetin, tangeretin, and nobiletin on HGF/c-Met-mediated tumor invasion and metastasis. Among them, nobiletin markedly inhibited HGF-induced the abilities of the adhesion, invasion, and migration by cell-matrix adhesion assay and transwell-chamber invasion/migration assay under non-cytotoxic concentrations. Data also showed nobiletin inhibited HGF-induced cell scattering and cytoskeleton changed such as filopodia and lamellipodia. Furthermore, nobiletin could inhibit HGF-induced the membrane localization of phosphorylated c-Met, ERK2, and Akt, but not phosphorylated JNK1/2 and p38. Next, nobiletin significantly decreased the levels of phospho-ERK2 and phospho-Akt in ERK2 or Akt siRNA-transfected cells concomitantly with a marked reduction on cell invasion and migration. In conclusion, nobiletin attenuates HGF-induced HepG2 cells metastasis involving both ERK and PI3K/Akt pathways and are potentially useful as anti-metastatic agents for the treatment of hepatoma.
Collapse
Affiliation(s)
- Ming-Der Shi
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | |
Collapse
|
12
|
Hepatocyte growth factor, a determinant of airspace homeostasis in the murine lung. PLoS Genet 2013; 9:e1003228. [PMID: 23459311 PMCID: PMC3573081 DOI: 10.1371/journal.pgen.1003228] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Accepted: 11/23/2012] [Indexed: 12/02/2022] Open
Abstract
The alveolar compartment, the fundamental gas exchange unit in the lung, is critical for tissue oxygenation and viability. We explored hepatocyte growth factor (HGF), a pleiotrophic cytokine that promotes epithelial proliferation, morphogenesis, migration, and resistance to apoptosis, as a candidate mediator of alveolar formation and regeneration. Mice deficient in the expression of the HGF receptor Met in lung epithelial cells demonstrated impaired airspace formation marked by a reduction in alveolar epithelial cell abundance and survival, truncation of the pulmonary vascular bed, and enhanced oxidative stress. Administration of recombinant HGF to tight-skin mice, an established genetic emphysema model, attenuated airspace enlargement and reduced oxidative stress. Repair in the TSK/+ mouse was punctuated by enhanced akt and stat3 activation. HGF treatment of an alveolar epithelial cell line not only induced proliferation and scattering of the cells but also conferred protection against staurosporine-induced apoptosis, properties critical for alveolar septation. HGF promoted cell survival was attenuated by akt inhibition. Primary alveolar epithelial cells treated with HGF showed improved survival and enhanced antioxidant production. In conclusion, using both loss-of-function and gain-of-function maneuvers, we show that HGF signaling is necessary for alveolar homeostasis in the developing lung and that augmentation of HGF signaling can improve airspace morphology in murine emphysema. Our studies converge on prosurvival signaling and antioxidant protection as critical pathways in HGF–mediated airspace maintenance or repair. These findings support the exploration of HGF signaling enhancement for diseases of the airspace. The airspace compartment of the mammalian lung, comprised of spherical sacs termed alveoli, harbors the architecture, cellular composition, and molecular armamentarium to perform the critical function of gas exchange or oxygen uptake. Despite the necessity of this alveolar compartment for organismal viability, the mechanism by which alveoli are formed and maintained is obscure. Furthermore, no treatments are currently available that can regenerate the airspace once damaged. In this manuscript, we sought to determine whether hepatocyte growth factor, a cytokine with a functional armamentarium that subserves the critical events of alveolar formation (epithelial proliferation, migration, resistance from apoptosis and angiogenesis), could be an important mediator of alveolar formation and airspace maintenance. Our simple paradigm was that critical homeostatic pathways for the lung should operate both in lung formation and in lung maintenance/regeneration. Using an informative battery of mouse models and cell lines, we show that hepatocyte growth factor is a determinant of alveolar formation and that the enhancement of hepatocyte growth factor signaling can both protect and repair the airspace from pathologic airspace enlargement or emphysema.
Collapse
|
13
|
Lin HC, Lai PY, Lin YP, Huang JY, Yang BC. Fas ligand enhances malignant behavior of tumor cells through interaction with Met, hepatocyte growth factor receptor, in lipid rafts. J Biol Chem 2012; 287:20664-73. [PMID: 22535954 DOI: 10.1074/jbc.m111.326058] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Many late-stage cancer cells express Fas ligand (FasL) and show high malignancy with metastatic potential. We report here a novel signaling mechanism for FasL that hijacks the Met signal pathway to promote tumor metastasis. FasL-expressing human tumor cells express a significant amount of phosphorylated Met. The down-regulation of FasL in these cells led to decreased Met activity and reduced cell motility. Ectopic expression of human FasL in NIH3T3 cells significantly stimulated their migration and invasion. The inhibition of Met and Stat3 activities reverted the FasL-associated phenotype. Notably, FasL variants activated the Met pathway, even though most of their intracellular domain or Fas binding sites were deleted. FasL interacted with Met through the FasL(105-130) extracellular region in lipid rafts, which consequently led to Met activation. Knocking down Met gene expression by RNAi technology reverted the FasL-associated motility to basal levels. Furthermore, treatment with synthetic peptides corresponding to FasL(117-126) significantly reduced the FasL/Met interaction, Met phosphorylation, and cell motility of FasL(+) transfectants and tumor cells. Finally, the transfectants of truncated FasL showed strong anchorage-independent growth and lung metastasis potential in null mice. Collectively, our results establish the FasL-Met-Stat3 signaling pathway and explains the metastatic phenotype of FasL-expressing tumors.
Collapse
Affiliation(s)
- Huan-Ching Lin
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | | | | | | | | |
Collapse
|
14
|
Honda K, Okamoto K, Mochida Y, Ishioka K, Oka M, Maesato K, Ikee R, Moriya H, Hidaka S, Ohtake T, Doi K, Fujita T, Kobayashi S, Noiri E. A novel mechanism in maggot debridement therapy: protease in excretion/secretion promotes hepatocyte growth factor production. Am J Physiol Cell Physiol 2011; 301:C1423-30. [PMID: 21881000 DOI: 10.1152/ajpcell.00065.2011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Maggot debridement therapy (MDT) is effective for treating intractable wounds, but its precise molecular mechanism, including the association between MDT and growth factors, remains unknown. We administered MDT to nine patients (66.3 ± 11.8 yr, 5 male and 4 female) with intractable wounds of lower extremities because they did not respond to conventional therapies. Significant increases of hepatocyte growth factor (HGF) levels were observed in femoral vein blood during 48 h of MDT (P < 0.05), but no significant change was found for vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), transforming growth factor-β1 (TGF-β1), or tumor necrosis factor-α (TNF-α). We conducted NIH-3T3 cell stimulation assay to evaluate the relation between HGF and protease activity in excretion/secretion (ES) derived from maggots. Compared with the control group, HGF was significantly higher in the 0.05 μg/ml ES group (P < 0.01). Furthermore, protease inhibitors suppressed the increase of HGF (P < 0.05). The HGF expression was increased in proportion to the ES protein concentration of 0.025 to 0.5 μg/ml. In fact, ES showed stronger capability of promoting HGF production and less cytotoxicity than chymotrypsin or bromelain. HGF is an important factor involved in cutaneous wound healing. Therefore, these results suggest that formation of healthy granulation tissue observed during MDT results from the increased HGF. Further investigation to identify molecules enhancing HGF expression by MDT will contribute greatly to drug target discovery for intractable wound healing therapy.
Collapse
Affiliation(s)
- Kenjiro Honda
- 107 Laboratory, Departments of Nephrology and Endocrinology, University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Syed ZA, Yin W, Hughes K, Gill JN, Shi R, Clifford JL. HGF/c-met/Stat3 signaling during skin tumor cell invasion: indications for a positive feedback loop. BMC Cancer 2011; 11:180. [PMID: 21595927 PMCID: PMC3112164 DOI: 10.1186/1471-2407-11-180] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2010] [Accepted: 05/19/2011] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Stat3 is a cytokine- and growth factor-inducible transcription factor that regulates cell motility, migration, and invasion under normal and pathological situations, making it a promising target for cancer therapeutics. The hepatocyte growth factor (HGF)/c-met receptor tyrosine kinase signaling pathway is responsible for stimulation of cell motility and invasion, and Stat3 is responsible for at least part of the c-met signal. METHODS We have stably transfected a human squamous cell carcinoma (SCC) cell line (SRB12-p9) to force the expression of a dominant negative form of Stat3 (S3DN), which we have previously shown to suppress Stat3 activity. The in vitro and in vivo malignant behavior of the S3DN cells was compared to parental and vector transfected controls. RESULTS Suppression of Stat3 activity impaired the ability of the S3DN cells to scatter upon stimulation with HGF (c-met ligand), enhanced their adhesion, and diminished their capacity to invade in vitro and in vivo. Surprisingly, S3DN cells also showed suppressed HGF-induced activation of c-met, and had nearly undetectable basal c-met activity, as revealed by a phospho-specific c-met antibody. In addition, we showed that there is a strong membrane specific localization of phospho-Stat3 in the wild type (WT) and vector transfected control (NEO4) SRB12-p9 cells, which is lost in the S3DN cells. Finally, co-immunoprecipitation experiments revealed that S3DN interfered with Stat3/c-met interaction. CONCLUSION These studies are the first confirm that interference with the HGF/c-met/Stat3 signaling pathway can block tumor cell invasion in an in vivo model. We also provide novel evidence for a possible positive feedback loop whereby Stat3 can activate c-met, and we correlate membrane localization of phospho-Stat3 with invasion in vivo.
Collapse
Affiliation(s)
- Zanobia A Syed
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center-Shreveport and Feist Weiller Cancer Center, Shreveport, Louisiana 71103, USA
| | | | | | | | | | | |
Collapse
|
16
|
Koh YW, Choi EC, Kang SU, Hwang HS, Lee MH, Pyun J, Park R, Lee Y, Kim CH. Green tea (-)-epigallocatechin-3-gallate inhibits HGF-induced progression in oral cavity cancer through suppression of HGF/c-Met. J Nutr Biochem 2011; 22:1074-83. [PMID: 21292466 DOI: 10.1016/j.jnutbio.2010.09.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 09/04/2010] [Accepted: 09/09/2010] [Indexed: 10/18/2022]
Abstract
Hepatocyte growth factor (HGF) and c-Met have recently attracted a great deal of attention as prognostic indicators of patient outcome, and they are important in the control of tumor growth and invasion. Epigallocatechin-3-gallate (EGCG) has been shown to modulate multiple signal pathways in a manner that controls the unwanted proliferation and invasion of cells, thereby imparting cancer chemopreventive and therapeutic effects. In this study, we investigated the effects of EGCG in inhibiting HGF-induced tumor growth and invasion of oral cancer in vitro and in vivo. We examined the effects of EGCG on HGF-induced cell proliferation, migration, invasion, induction of apoptosis and modulation of HGF/c-Met signaling pathway in the KB oral cancer cell line. We investigated the antitumor effect and inhibition of c-Met expression by EGCG in a syngeneic mouse model (C3H/HeJ mice, SCC VII/SF cell line). HGF promoted cell proliferation, migration, invasion and induction of MMP (matrix metalloproteinase)-2 and MMP-9 in KB cells. EGCG significantly inhibited HGF-induced phosphorylation of Met and cell growth, invasion and expression of MMP-2 and MMP-9. EGCG blocked HGF-induced phosphorylation of c-Met and that of the downstream kinases AKT and ERK, and inhibition of p-AKT and p-ERK by EGCG was associated with marked increases in the phosphorylation of p38, JNK, cleaved caspase-3 and poly-ADP-ribose polymerase. In C3H/HeJ syngeneic mice, as an in vivo model, tumor growth was suppressed and apoptosis was increased by EGCG. Our results suggest that EGCG may be a potential therapeutic agent to inhibit HGF-induced tumor growth and invasion in oral cancer.
Collapse
Affiliation(s)
- Yoon Woo Koh
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Wohlmann A, Sebastian K, Borowski A, Krause S, Friedrich K. Signal transduction by the atopy-associated human thymic stromal lymphopoietin (TSLP) receptor depends on Janus kinase function. Biol Chem 2010; 391:181-186. [PMID: 20128689 DOI: 10.1515/bc.2010.029] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Thymic stromal lymphopoietin (TSLP) is an interleukin-(IL)-7-like cytokine with emerging pathological importance for the development of atopic diseases such as allergic asthma bronchiale. The TSLP receptor (TSLPR), a heterodimeric type I cytokine receptor, shares the IL-7R alpha-subunit with the IL-7 receptor system. The specific TSLPR alpha-chain shows similarities with the gammac receptor chain, but has some unusual features within the receptor family in both its ligand-binding and cytoplasmic domain. The murine TSLPR signals via the signal transducers and activators of transcription STAT5 and STAT3, but is unique among cytokine receptors in that it activates STATs without the involvement of Janus (JAK) tyrosine kinases, but instead utilizes the Src type kinase Tec. Here, we show by Western blotting and reporter gene experiments in combination with the application of a specific JAK inhibitor that the human TSLP receptor, in contrast, requires the function of JAK1 and JAK2 for STAT activation. Moreover, we demonstrate that the human TSLPR mediates gene regulation not only through STAT5 and STAT3 but has also the potential to mediate transcription via STAT1. Our work should help to understand more thoroughly how TSLP triggers inflammatory responses in the course of atopic diseases.
Collapse
Affiliation(s)
- Andreas Wohlmann
- Institute of Biochemistry II, University of Jena Medical School, Nonnenplan 2, D-07743 Jena, Germany
| | - Katrin Sebastian
- Institute of Biochemistry II, University of Jena Medical School, Nonnenplan 2, D-07743 Jena, Germany
| | - Andreas Borowski
- Institute of Biochemistry II, University of Jena Medical School, Nonnenplan 2, D-07743 Jena, Germany
| | - Sebastian Krause
- Institute of Biochemistry II, University of Jena Medical School, Nonnenplan 2, D-07743 Jena, Germany
| | - Karlheinz Friedrich
- Institute of Biochemistry II, University of Jena Medical School, Nonnenplan 2, D-07743 Jena, Germany
| |
Collapse
|
18
|
Qian J, Zhu CH, Tang S, Shen AJ, Ai J, Li J, Geng MY, Ding J. alpha2,6-hyposialylation of c-Met abolishes cell motility of ST6Gal-I-knockdown HCT116 cells. Acta Pharmacol Sin 2009; 30:1039-45. [PMID: 19483716 DOI: 10.1038/aps.2009.84] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
AIM We aimed to investigate the potential modification of previously unrecognized surface glycoprotein(s) by alpha2,6-sialylation other than by integrins. METHODS The expression of beta-galactoside alpha2,6-sialyltransferase (ST6Gal-I) in the colon cancer cell line HCT116 was reduced by siRNA. The adhesion and Boyden chamber assay were used to detect the variation in cell motility. alpha2,6-Sialylation proteins were detected with lectin affinity assay. The mRNA expression, protein expression and downstream signaling modulation with siRNA were detected using reverse transcription-polymerase chain reaction, flow cytometry analysis, and Western blot. RESULTS In HCT116 cells, the knockdown of ST6Gal-I inhibited cell motility, but did not affect cell adhesion. This selectively altered cell migration was caused by the loss of alpha2,6-sialic acid structures on c-Met. Moreover, STAT3 was dephosphorylated at tyrosine 705 in ST6Gal-I-knockdown (ST6Gal-I-KD) HCT116 cells. CONCLUSION c-Met is the substrate of ST6Gal-I. The hyposialylation of c-Met can abolish cell motility in ST6Gal-I-KD HCT116 cells.Acta Pharmacologica Sinica (2009) 30: 1039-1045; doi: 10.1038/aps.2009.84; published online 1 June 2009.
Collapse
|
19
|
Lee BS, Park M, Cha HY, Lee JH. Hepatocyte growth factor induces delayed STAT3 phosphorylation through interleukin-6 expression. Cell Signal 2008; 21:419-27. [PMID: 19071214 DOI: 10.1016/j.cellsig.2008.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2008] [Revised: 11/13/2008] [Accepted: 11/15/2008] [Indexed: 11/30/2022]
Abstract
Met receptor tyrosine kinase mediates pleiotropic cellular responses following its activation by hepatocyte growth factor or scatter factor (HGF/SF). STAT3 was reported to be one of direct downstream molecules in HGF/SF-Met signaling. In the present study, however, we observed that Tyr705 of STAT3 was phosphorylated from 2 h or 6 h in NIH3T3 and Chang liver cells, respectively, after HGF/SF treatment. Blocking of the phosphorylation by cycloheximide or actinomycin D and the rapid STAT3 phosphorylation with the conditioned medium from HGF/SF-treated NIH3T3 cells suggested that a newly synthesized secretory protein was responsible for the delayed STAT3 phosphorylation. Among the known mediators to induce STAT3 phosphorylation, interleukin-6 (IL-6) mRNA and protein were induced by HGF/SF, and the released IL-6 was accumulated in the conditioned medium after HGF/SF treatment. Furthermore, the neutralizing IL-6 antibody abolished the STAT3 phosphorylation. Treatment with LY294002, a PI3 kinase inhibitor, but not with other signal inhibitors, resulted in the loss of delayed STAT3 phosphorylation by HGF/SF, showing the involvement of PI3 kinase pathway. Collectively, these results demonstrate that HGF/SF-Met signal cascade stimulates IL-6 production via PI3 kinase pathway, leading to STAT3 phosphorylation as a secondary effect.
Collapse
Affiliation(s)
- Bok-Soon Lee
- Department of Biochemistry and Molecular Biology, Ajou University Medical School, 5 Wonchon-Dong, Yeongtong-Gu, Suwon 443-721, South Korea
| | | | | | | |
Collapse
|
20
|
Kermorgant S, Parker PJ. Receptor trafficking controls weak signal delivery: a strategy used by c-Met for STAT3 nuclear accumulation. ACTA ACUST UNITED AC 2008; 182:855-63. [PMID: 18779368 PMCID: PMC2528569 DOI: 10.1083/jcb.200806076] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
C-Met, the receptor of hepatocyte growth factor (HGF), through overexpression or mutation, is a major protooncogene that provides an attractive molecular target for cancer therapy. HGF/c-Met-induced tumorigenesis is dependent, in part, on the transcription factor and oncogene signal transducer and activator of transcription 3 (STAT3), which is believed to be activated by the receptor at the plasma membrane and then to travel to the nucleus where it acts. We demonstrate that although the robust signal to STAT3 elicited from the cytokine oncostatin-M does indeed support this mechanism of STAT3 action, for the weaker STAT3 signal emanating from c-Met, the activated receptor itself needs to be delivered to a perinuclear endosomal compartment to sustain phosphorylated STAT3 in the nucleus. This is signal specific because c-Met-induced extracellular signal-regulated kinase nuclear accumulation does not require receptor trafficking to the perinuclear compartment. This response is triggered from peripheral endosomes. Thus, control of growth factor receptor traffic determines the nature of the signal output, providing novel opportunities for intervention.
Collapse
Affiliation(s)
- Stéphanie Kermorgant
- Department of Tumour Biology, Cancer Research UK Clinical Centre, Bart's and the London Queen Mary's School of Medicine and Dentistry, London EC1M 6BQ, England, UK
| | | |
Collapse
|
21
|
Takahashi M, Sugiura T, Abe M, Ishii K, Shirasuna K. Regulation of c-Met signaling by the tetraspanin KAI-1/CD82 affects cancer cell migration. Int J Cancer 2007; 121:1919-1929. [PMID: 17621632 DOI: 10.1002/ijc.22887] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
It has been proposed that the metastasis suppressor CD82/KAI-1, which is a member of the tetraspanin superfamily, regulates biological activity by associating with cell surface receptors or proteins. We show a novel association between CD82 and the hepatocyte growth factor (HGF) receptor c-Met. Although ectopic expression of CD82 in nonsmall cell lung carcinoma cells did not affect the tyrosine phosphorylation of c-Met, these cells showed significant suppression of HGF-induced lamellipodial protrusion and cell migration. CD82 selectively attenuated c-Met signaling via the Ras-Cdc42/Rac and the phosphatidylinositol 3-kinase/Cdc42/Rac pathways. In contrast, another c-Met signaling pathway that involves phosphatidylinositol 3-kinase/Akt and phosphatidylinositol 3-kinase/mitogen activated protein kinase was not affected by CD82. Signaling adapter proteins for c-Met, such as Grb2 and p85, exhibited reduced association with c-Met in cells that ectopically expressed CD82. These results indicate that the CD82-c-Met complex inhibits HGF-induced cancer cell migration by the inactivation of small GTP-binding proteins of the Rho family via c-Met adapter proteins.
Collapse
Affiliation(s)
- Miho Takahashi
- Department of Oral and Maxillofacial Surgery, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Tsuyoshi Sugiura
- Department of Oral and Maxillofacial Surgery, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masakazu Abe
- Department of Oral and Maxillofacial Surgery, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Koutaro Ishii
- Department of Oral and Maxillofacial Surgery, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Kanemitsu Shirasuna
- Department of Oral and Maxillofacial Surgery, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Tsareva SA, Moriggl R, Corvinus FM, Wiederanders B, Schütz A, Kovacic B, Friedrich K. Signal transducer and activator of transcription 3 activation promotes invasive growth of colon carcinomas through matrix metalloproteinase induction. Neoplasia 2007; 9:279-91. [PMID: 17460772 PMCID: PMC1854849 DOI: 10.1593/neo.06820] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 02/28/2007] [Accepted: 03/02/2007] [Indexed: 12/20/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is aberrantly activated in colorectal carcinomas (CRCs). Here, we define the relationship between STAT3 function and the malignant properties of colon carcinoma cells. Elevated activation of STAT3 enhances invasive growth of the CRC cell lines. To address mechanisms through which STAT3 influences invasiveness, the protease mRNA expression pattern of CRC biopsies was analyzed and correlated with the STAT3 activity status. These studies revealed a striking coincidence of STAT3 activation and strong expression of matrix metalloproteinases MMP-1, -3, -7, and -9. Immunohistological examination of CRC tumor specimens showed a clear colocalization of MMP-1 and activated STAT3. Experimentally induced STAT3 activity in CRC cell lines enhanced both the level of MMP-1 mRNA and secreted MMP-1 enzymatic activity. A direct connection of STAT3 activity and transcription from the MMP-1 promoter was shown by reporter gene experiments. Moreover, high-affinity binding of STAT3 to STAT recognition elements in both the MMP-1 and MMP-3 promoter was demonstrated. Xenograft tumors arising from implantation of CRC cells into nude mice showed simultaneous appearance and colocalization of p-Y-STAT3 and MMP-1 expression. Our results link aberrant activity of STAT3 in CRC to malignant tumor progression through upregulated expression of MMPs.
Collapse
Affiliation(s)
- Svetlana A Tsareva
- Institute of Biochemistry I, Friedrich-Schiller University Jena Medical School, Jena, Germany
- Novosibirsk State Medical Academy, Novosibirsk, Russia
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Florian M Corvinus
- Institute of Biochemistry I, Friedrich-Schiller University Jena Medical School, Jena, Germany
| | - Bernd Wiederanders
- Institute of Biochemistry I, Friedrich-Schiller University Jena Medical School, Jena, Germany
| | | | - Boris Kovacic
- Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Karlheinz Friedrich
- Institute of Biochemistry I, Friedrich-Schiller University Jena Medical School, Jena, Germany
| |
Collapse
|
23
|
Tsuda M, Davis IJ, Argani P, Shukla N, McGill GG, Nagai M, Saito T, Laé M, Fisher DE, Ladanyi M. TFE3 fusions activate MET signaling by transcriptional up-regulation, defining another class of tumors as candidates for therapeutic MET inhibition. Cancer Res 2007; 67:919-29. [PMID: 17283122 DOI: 10.1158/0008-5472.can-06-2855] [Citation(s) in RCA: 240] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Specific chromosomal translocations encoding chimeric transcription factors are considered to play crucial oncogenic roles in a variety of human cancers but the fusion proteins themselves seldom represent suitable therapeutic targets. Oncogenic TFE3 fusion proteins define a subset of pediatric renal adenocarcinomas and one fusion (ASPL-TFE3) is also characteristic of alveolar soft part sarcoma (ASPS). By expression profiling, we identified the MET receptor tyrosine kinase gene as significantly overexpressed in ASPS relative to four other types of primitive sarcomas. We therefore examined MET as a direct transcriptional target of ASPL-TFE3. ASPL-TFE3 binds to the MET promoter and strongly activates it. Likewise, PSF-TFE3 and NONO-TFE3 also bind this promoter. Induction of MET by ASPL-TFE3 results in strong MET autophosphorylation and activation of downstream signaling in the presence of hepatocyte growth factor (HGF). In cancer cell lines containing endogenous TFE3 fusion proteins, inhibiting MET by RNA interference or by the inhibitor PHA665752 abolishes HGF-dependent MET activation, causing decreased cell growth and loss of HGF-dependent phenotypes. MET is thus a potential therapeutic target in these cancers. Aberrant transcriptional up-regulation of MET by oncogenic TFE3 fusion proteins represents another mechanism by which certain cancers become dependent on MET signaling. The identification of kinase signaling pathways transcriptionally up-regulated by oncogenic fusion proteins may reveal more accessible therapeutic targets in this class of human cancers.
Collapse
Affiliation(s)
- Masumi Tsuda
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Populations of self-renewing cells that arise during normal embryonic development harbour the potential for rapid proliferation, migration or transdifferentiation and, therefore, tumour generation. So, control mechanisms are essential to prevent rapidly expanding populations from malignant growth. Transcription factors have crucial roles in ensuring establishment of such regulation, with the Pax gene family prominent amongst these. This review examines the role of Pax family members during embryogenesis, and their contribution to tumorigenesis when subverted.
Collapse
Affiliation(s)
- Ewan J D Robson
- Department of Pathology, University of Otago, PO Box 913, Dunedin 9001, New Zealand
| | | | | |
Collapse
|