1
|
Zhao H, Martin E, Matalkah F, Shah N, Ivanov A, Ruppert JM, Lockman PR, Agazie YM. Conditional knockout of SHP2 in ErbB2 transgenic mice or inhibition in HER2-amplified breast cancer cell lines blocks oncogene expression and tumorigenesis. Oncogene 2019; 38:2275-2290. [PMID: 30467378 PMCID: PMC6440805 DOI: 10.1038/s41388-018-0574-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 09/13/2018] [Accepted: 10/04/2018] [Indexed: 01/21/2023]
Abstract
Overexpression of the human epidermal growth factor receptor 2 (HER2) is the cause of HER2-positive breast cancer (BC). Although HER2-inactivating therapies have benefited BC patients, development of resistance and disease recurrence have been the major clinical problems, pointing to a need for alternative therapeutic strategies. For that to happen, proteins that play critical roles in the biology of HER2-induced tumorigenesis have to be identified and characterized. Here, we show that the Src homology phosphotyrosyl phosphatase 2 (Shp2) encoded by the Ptpn11 gene is a requisite for ErbB2-induced tumorigenesis. We report that conditional knockout of Shp2 alleles in the ErbB2 BC model mice abrogates mammary tumorigenesis by blocking the expression of the ErbB2 transgene. We also show that inhibition of SHP2 encoded by the PTPN11 gene in the HER2-amplified BC cells induces a normal-like cellular phenotype and suppresses tumorigenesis and metastasis by blocking HER2 overexpression. These findings demonstrate that ErbB2-induced tumors in mice or xenograft tumors induced by transplantation of HER2-amplified BC cells are vulnerable to SHP2 inhibition since it abrogates the expression of the very oncogene that causes of the disease. This report paves the way for developing SHP2-targeting therapies for BC treatment in the future.
Collapse
Affiliation(s)
- Hua Zhao
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Elisha Martin
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Fatimah Matalkah
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Neal Shah
- Department of Basic Pharmaceutical Sciences, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Alexey Ivanov
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - J Michael Ruppert
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Paul R Lockman
- Department of Basic Pharmaceutical Sciences, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Yehenew M Agazie
- Department of Biochemistry, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA.
- WVU Cancer Institute, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
2
|
Chen MJ, Wang YC, Wu DW, Chen CY, Lee H. Association of nuclear localization of SHP2 and YAP1 with unfavorable prognosis in non-small cell lung cancer. Pathol Res Pract 2019; 215:801-806. [PMID: 30685130 DOI: 10.1016/j.prp.2019.01.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/02/2019] [Accepted: 01/17/2019] [Indexed: 01/07/2023]
Abstract
Src homology region 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2) is ubiquitously expressed in cytoplasmic localization, which in turn confers tumor malignancy and poor prognosis in various human cancers. YAP1 interacts with SHP2 to promote translocation of SHP2 to nucleus, which consequently promotes Wnt target activation. However, the oncogenic role of the nuclear localization of SHP2 in human cancers remains unclear. We hypothesized that nuclear SHP2 localization, in combination with nuclear YAP1 expression, could be associated with poor overall survival (OS) and relapse free survival (RFS) due to an increase in cyclin D1 and c-Myc mRNA expression following activation of Wnt/ß-catenin signaling. Immunohistochemical analysis of SHP2 and YAP1 protein expression in 102 tumors resected from patients with NSCLC revealed that nuclear SHP2 expression was well correlated with nuclear YAP1 expression (P < 0.001). Evaluation of cyclin D1 and c-Myc mRNA levels by the real-time reverse-phase polymerase chain reaction (RT-PCR) revealed that patients with high cyclin D1 and high c-Myc mRNA expressing tumors more commonly showed high nuclear YAP1 and high nuclear SHP2 (high/high) rather than the high/low, low/high, or low/low combinations (P < 0.001 for cyclin D1 and c-Myc). Kaplan-Meier and Cox-regression models showed OS and RFS to be poorer in patients in the high/high subgroup than in the low/low subgroup (OS: HR = 2.85, 95% CI, 1.52-5.35, P = 0.001; RFS: HR = 2.55, 95% CI, 1.37-4.72, P = 0.003). No prognostic significance was observed for the other two subgroups (low/high and high/low) when compared to the low/low subgroup in this study population. Therefore, we suggest that the prognostic value of SHP2 could reflect the nuclear localization of SHP2 and its interaction with nuclear YAP1, which led to subsequent upregulation of cyclin D1 and c-Myc mRNA expression via activation of the Wnt/ß-catenin signaling pathway.
Collapse
Affiliation(s)
- Ming-Jenn Chen
- Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan; Department of Sports Management, College of Leisure and Recreation Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan.
| | - Yao-Chen Wang
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - De-Wei Wu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Chi-Yi Chen
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
3
|
Dong Y, Chang C, Liu J, Qiang J. Targeting of GIT1 by miR-149* in breast cancer suppresses cell proliferation and metastasis in vitro and tumor growth in vivo. Onco Targets Ther 2017; 10:5873-5882. [PMID: 29270025 PMCID: PMC5729835 DOI: 10.2147/ott.s144280] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Breast cancer remains a major cause of cancer-related death in women worldwide. Dysregulation of microRNAs (miRNAs) is involved in the initiation and progression of breast cancer. Moreover, it was found that GIT1 was widely involved in the development of many human cancers. Herein, we aimed to investigate the expression changes of miR-149* and GIT1 and the functional effects of miR-149*/GIT1 link in breast cancer. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot (WB) were used to examine the expression levels of miR-149* and GIT1. Dual luciferase reporter assay was utilized to confirm the target interaction between miR-149* and GIT1. The biological functions, including cell proliferation, invasion, and migration, of miR-149* and GIT1 were determined by MTT assay and Transwell assays, respectively. Eventually, the tumor xenograft model in nude mice injected with stable transfected MDA-MB-231 cells was established to verify the effects of miR-149* and GIT1 on tumor growth. Our results showed that miR-149* expression was decreased, whereas GIT1 expression was increased in clinical samples of breast cancer. Based on the inverse expression trend between miR-149* and GIT1, we further demonstrated that miR-149* indeed directly targets GIT1. Subsequently, it was observed that inhibition of miR-149* significantly promoted cell proliferation, invasion, and migration, but the ability of cell proliferation, invasion, and migration was obviously declined after silencing of GIT1 in MDA-MB-231 cells transfected with miR-149* mimic and/or si-GIT1. Finally, it was also found that elevated miR-149* decelerated the tumor growth, while restored GIT1 accelerated the tumor growth in nude mice after 35 days of tumor xenograft. Collectively, these findings concluded that miR-149* might exert a tumor suppressive role in breast cancer by targeting GIT1.
Collapse
Affiliation(s)
- Yan Dong
- Department of Ultrasonography, Jinshan Hospital
| | - Cai Chang
- Department of Ultrasonography, Cancer Center
| | | | - Jinwei Qiang
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Rong L, Zhou S, Liu X, Li A, Jing T, Liu X, Zhang Y, Cai S, Tang X. Trastuzumab-modified DM1-loaded nanoparticles for HER2 + breast cancer treatment: an in vitro and in vivo study. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1708-1718. [PMID: 29069935 DOI: 10.1080/21691401.2017.1391821] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Emtansine (DM1) is a highly potent anti-microtubule agent that has shown promising results for breast cancer treatment, but side effects limit its widespread clinical use. In this research, a new nano-drug was developed to integrate DM1 agent with antibody targeting. METHODS A system of novel nanoparticles (NPs) DM1-NPs-trastuzumab (DM1-NPs-Tmab) of DM1 combined with (anti-HER2 antibody, Herceptin®, Trastuzumab) was developed for HER2+ breast cancer treatment, and its physical characterization and antitumor biological activity were investigated. RESULTS DM1-NPs-Tmab-targeted HER2+ breast cancer cells specifically were developed. Compared with naked DM1 and Herceptin, DM1-NPs-Tmab showed greater toxicity on HER2+ cancer cells and blocked the HER2-PI3K/Akt cell activation pathway. DM1-NPs-Tmab inhibited tumor growth by 88% and had less toxic effects in vivo than non-targeting DM1 when administered to MDA-MB-453 xenograft bearing mice. CONCLUSION DM1-NPs-Tmab shows superior anti-tumor efficacy than free Herceptin or DM1. DM1-NPs-Tmab is a potential promising formulation for targeting biotherapy of HER2+ tumors.
Collapse
Affiliation(s)
- Ling Rong
- a Bozhou People's Hospital Affiliated to Medical College , Anhui University of Science & Technology , Bozhou , China.,b Medical College , Anhui University of Science & Technology , Huainan , China
| | - Shuping Zhou
- c Huainan First People's Hospital and First Affiliated Hospital of Medical College , Anhui University of Science & Technology , Huainan , China
| | - Xinkuang Liu
- c Huainan First People's Hospital and First Affiliated Hospital of Medical College , Anhui University of Science & Technology , Huainan , China
| | - Amin Li
- c Huainan First People's Hospital and First Affiliated Hospital of Medical College , Anhui University of Science & Technology , Huainan , China
| | - Tao Jing
- b Medical College , Anhui University of Science & Technology , Huainan , China
| | - Xueke Liu
- b Medical College , Anhui University of Science & Technology , Huainan , China
| | - Yinci Zhang
- b Medical College , Anhui University of Science & Technology , Huainan , China
| | - Shiyu Cai
- b Medical College , Anhui University of Science & Technology , Huainan , China
| | - Xiaolong Tang
- a Bozhou People's Hospital Affiliated to Medical College , Anhui University of Science & Technology , Bozhou , China.,b Medical College , Anhui University of Science & Technology , Huainan , China
| |
Collapse
|
5
|
Yang X, Tang C, Luo H, Wang H, Zhou X. Shp2 confers cisplatin resistance in small cell lung cancer via an AKT-mediated increase in CA916798. Oncotarget 2017; 8:23664-23674. [PMID: 28423588 PMCID: PMC5410335 DOI: 10.18632/oncotarget.15641] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 12/05/2016] [Indexed: 01/09/2023] Open
Abstract
The tyrosine phosphatase Shp2 is associated with tumorigenesis in small cell lung cancer (SCLC). However, the relationship between Shp2 and resistance to chemotherapy remains unclear. Here, we show that Shp2 plays an important role in inducing resistance to cisplatin-based chemotherapy via the SHP2-AKT-CA916798 pathway. In an SCLC cell line, overexpression of Shp2 induced cisplatin resistance and the increased expression of AKT, pAKT, pmTOR, and CA916798. Conversely, depletion of Shp2 in a cisplatin-resistant cell line via RNA interference increased cisplatin sensitivity and decreased AKT, pAKT, pmTOR, and CA916798 expression levels. Activation of AKT stimulated CA916798 expression and altered the level of Shp2. A mouse xenograft model verified the results obtained from the in vitro experiments. In addition, we collected and analyzed clinical SCLC specimens and found that Shp2 levels correlated with CA916798 expression in tumor tissues. Importantly, higher levels of Shp2 or CA916798 were associated with a poorer prognosis in SCLC patients who received chemotherapy. Together, our findings indicate that Shp2 induces cisplatin resistance in SCLC patients via the SHP2-AKT-CA916798 pathway. Therefore, Shp2 and CA916798 may be promising biomarkers for predicting resistance to chemotherapy and may function as targets for enhancing treatments.
Collapse
Affiliation(s)
- Xuemei Yang
- Department of Respiratory, Southwest Hospital, Third Military Medical University, Shapingba District, Chongqing 400038, PR China.,Department of Respiratory, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Yuzhong District, Chongqing 400042, PR China
| | - Chunlan Tang
- Department of Respiratory, Southwest Hospital, Third Military Medical University, Shapingba District, Chongqing 400038, PR China
| | - Hu Luo
- Department of Respiratory, Southwest Hospital, Third Military Medical University, Shapingba District, Chongqing 400038, PR China
| | - Haijing Wang
- Department of Respiratory, Southwest Hospital, Third Military Medical University, Shapingba District, Chongqing 400038, PR China
| | - Xiangdong Zhou
- Department of Respiratory, Southwest Hospital, Third Military Medical University, Shapingba District, Chongqing 400038, PR China
| |
Collapse
|
6
|
Elson A. Stepping out of the shadows: Oncogenic and tumor-promoting protein tyrosine phosphatases. Int J Biochem Cell Biol 2017; 96:135-147. [PMID: 28941747 DOI: 10.1016/j.biocel.2017.09.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 12/18/2022]
Abstract
Protein tyrosine phosphorylation is critical for proper function of cells and organisms. Phosphorylation is regulated by the concerted but generically opposing activities of tyrosine kinases (PTKs) and tyrosine phosphatases (PTPs), which ensure its proper regulation, reversibility, and ability to respond to changing physiological situations. Historically, PTKs have been associated mainly with oncogenic and pro-tumorigenic activities, leading to the generalization that protein dephosphorylation is anti-oncogenic and hence that PTPs are tumor-suppressors. In many cases PTPs do suppress tumorigenesis. However, a growing body of evidence indicates that PTPs act as dominant oncogenes and drive cell transformation in a number of contexts, while in others PTPs support transformation that is driven by other oncogenes. This review summarizes the known transforming and tumor-promoting activities of the classical, tyrosine specific PTPs and highlights their potential as drug targets for cancer therapy.
Collapse
Affiliation(s)
- Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
7
|
Wang Q, Yang ZL, Zou Q, Yuan Y, Li J, Liang L, Zeng G, Chen S. SHP2 and UGP2 are Biomarkers for Progression and Poor Prognosis of Gallbladder Cancer. Cancer Invest 2016; 34:255-64. [DOI: 10.1080/07357907.2016.1193745] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
8
|
Matalkah F, Martin E, Zhao H, Agazie YM. SHP2 acts both upstream and downstream of multiple receptor tyrosine kinases to promote basal-like and triple-negative breast cancer. Breast Cancer Res 2016; 18:2. [PMID: 26728598 PMCID: PMC4700603 DOI: 10.1186/s13058-015-0659-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/25/2015] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Dysregulated receptor tyrosine kinase (RTK) signaling is a common occurrence in basal-like and triple-negative breast cancer (BTBC). As a result, RTK-targeting therapies have been initiated but proved difficult, mainly owing to the multiplicity of dysregulated RTKs. Hence, targeting master regulators of RTK signaling might alleviate this obstacle. Before that, however, defining the mechanism of such molecules is required. In this report, we show that the Src homology phosphotyrosyl phosphatase 2 (SHP2) is a master regulator of RTK expression and signaling in BTBC. METHODS Xenograft tumor growth studies were used to determine the effect of SHP2 inhibition on tumorigenesis and/or metastasis. Cell proliferation rate, anchorage-independent growth, mammosphere formation, and ALDEFLUOR assays were used to compare the relative functional importance of SHP2 and the epidermal growth factor receptor (EGFR) in BTBC cells. Immunohistochemistry and immunofluorescence analyses were used to determine the state of SHP2 and EGFR coexpression in BTBC. Analysis of mitogenic and cell survival signaling was performed to show SHP2's role in signaling by multiple RTKs. RESULTS Inhibition of SHP2 in BTBC cells suppresses their tumorigenic and metastatic properties. Because EGFR is the most commonly dysregulated RTK in BTBC, we first tested the effect of SHP2 inhibition on EGFR signaling and found that SHP2 is important not only for mediation of the Ras/extracellular signal-regulated kinase and the phosphatidyl inositol 3-kinase/Akt signaling pathways but also for the expression of the receptor itself. The existence of a tight association between SHP2 and EGFR expression in tumors and cell lines further suggested the importance of SHP2 in EGFR expression. Comparison of relative biological significance showed the superiority of SHP2 inhibition over that of EGFR, suggesting the existence of additional RTKs regulated by SHP2. Indeed, we found that the expression as well as the signaling efficiency of c-Met and fibroblast growth factor receptor 1, two other RTKs known to be dysregulated in BTBC, are SHP2-dependent. To our knowledge, this is the first demonstration of SHP2 acting both upstream and downstream of RTKs to promote signaling. CONCLUSIONS SHP2 upregulates the expression and signaling of multiple RTKs to promote BTBC. These findings provide a mechanistic explanation for the superiority of SHP2 inhibition in BTBC.
Collapse
Affiliation(s)
- Fatimah Matalkah
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Elisha Martin
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Hua Zhao
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Yehenew M Agazie
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA. .,The Marry Babb Randolph Cancer Center, School of Medicine, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
9
|
Zheng J, Huang S, Huang Y, Song L, Yin Y, Kong W, Chen X, Ouyang X. Expression and prognosis value of SHP2 in patients with pancreatic ductal adenocarcinoma. Tumour Biol 2015; 37:7853-9. [PMID: 26695153 DOI: 10.1007/s13277-015-4675-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/16/2015] [Indexed: 12/14/2022] Open
Abstract
SHP2 is an src homology (SH) 2 domain-containing protein tyrosine phosphatase (PTP). SHP2 implicitly contributes to tumorigenesis, but the role of SHP2 in pancreatic ductal adenocarcinoma is still unknown. The purpose of this study was to evaluate the prognostic significance and associated expression of SHP2 in pancreatic ductal adenocarcinoma (PDAC) patients. We used immunohistochemistry to assess the protein expression levels of SHP2 in 79 PDAC specimens. The correlations between SHP2 expression and various clinicopathological features were evaluated by Pearson's chi-square (χ (2)) test, Fisher's exact test, and Spearman's rank. Univariate and multivariate Cox regression analyses were used to identify correlations between the immunohistochemical data for SHP2 expression and the clinicopathologic characteristics in PDAC. Kaplan-Meier survival analysis was used to demonstrate the relation between overall survival and the expression of SHP2. Immunohistochemistry revealed significantly higher rates of high SHP2 expression in PDAC tissues (55.7 %) versus adjacent non-cancer tissues (10.1 %) (P < 0.05). Expression of SHP2 was only significantly correlated with histological differentiation (P = 0.033) and vital status (P = 0.025). Patients with high SHP2 expression had shorter overall survival times compared to those with low SHP2 expression (P = 0.000). Multivariate Cox regression analysis revealed that SHP2 overexpression was an independent prognostic factor in PDAC (P = 0.012). Our study demonstrated for the first time that higher expression of SHP2 might be involved in the progression of pancreatic ductal adenocarcinoma, suggesting that SHP2 may be a potential prognostic marker and target for therapy.
Collapse
Affiliation(s)
- Jiawei Zheng
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fujian, China
| | - Shanshan Huang
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fujian, China
| | - Yufang Huang
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fujian, China
| | - Li Song
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fujian, China
| | - Yin Yin
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Medical College, Xiamen University, Xiamen, China
| | - Wencui Kong
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fujian, China
| | - Xiong Chen
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fujian, China.
| | - Xuenong Ouyang
- Department of Medical Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzong Clinical College, Fujian Medical University, Fujian, China
| |
Collapse
|
10
|
Lan L, Holland JD, Qi J, Grosskopf S, Rademann J, Vogel R, Györffy B, Wulf-Goldenberg A, Birchmeier W. Shp2 signaling suppresses senescence in PyMT-induced mammary gland cancer in mice. EMBO J 2015; 34:1493-508. [PMID: 25736378 DOI: 10.15252/embj.201489004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 02/04/2015] [Indexed: 12/26/2022] Open
Abstract
In this study, we have used techniques from cell biology, biochemistry, and genetics to investigate the role of the tyrosine phosphatase Shp2 in tumor cells of MMTV-PyMT mouse mammary glands. Genetic ablation or pharmacological inhibition of Shp2 induces senescence, as determined by the activation of senescence-associated β-gal (SA-β-gal), cyclin-dependent kinase inhibitor 1B (p27), p53, and histone 3 trimethylated lysine 9 (H3K9me3). Senescence induction leads to the inhibition of self-renewal of tumor cells and blockage of tumor formation and growth. A signaling cascade was identified that acts downstream of Shp2 to counter senescence: Src, focal adhesion kinase, and Map kinase inhibit senescence by activating the expression of S-phase kinase-associated protein 2 (Skp2), Aurora kinase A (Aurka), and the Notch ligand Delta-like 1 (Dll1), which block p27 and p53. Remarkably, the expression of Shp2 and of selected target genes predicts human breast cancer outcome. We conclude that therapies, which rely on senescence induction by inhibiting Shp2 or controlling its target gene products, may be useful in blocking breast cancer.
Collapse
Affiliation(s)
- Linxiang Lan
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Jane D Holland
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Jingjing Qi
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Stefanie Grosskopf
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | | | - Regina Vogel
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Balázs Györffy
- MTA TTK Lendület Cancer Biomarker Research Group, Budapest, Hungary 2 Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | | | - Walter Birchmeier
- Cancer Research Program, Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| |
Collapse
|
11
|
Cai P, Guo W, Yuan H, Li Q, Wang W, Sun Y, Li X, Gu Y. Expression and clinical significance of tyrosine phosphatase SHP-2 in colon cancer. Biomed Pharmacother 2013; 68:285-90. [PMID: 24439672 DOI: 10.1016/j.biopha.2013.10.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 10/28/2013] [Indexed: 11/25/2022] Open
Abstract
Protein-tyrosine phosphatase SHP-2, encoded by gene PTPN11, has been identified as a tumor-promoting factor in several types of leukemia and is hyper-activated by other mechanisms in some solid tumors including gastric cancer, breast cancer, non-small cell lung cancer (NSCLC), etc. But few were reported on the expression and significances of SHP-2 in colon cancer. Here, we detect SHP-2 expression in colon cancer cells, colon cancer-induced by AOM+DSS in mice and 232 human colon cancer specimens, including 58 groups of self-matched adjacent peritumor tissues and normal tissues. We found that compared to the normal colon tissues, SHP-2 significantly decreased in tumor tissues (P<0.001). The same results were got in colon tumor cells as well as mice colon tumors. And in humans samples, low SHP-2 expression showed a significantly correlation with poor tumor differentiation (P<0.05), late TNM stage (P=0.1666) and lymph node metastasis (P<0.05).
Collapse
Affiliation(s)
- Peifen Cai
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China; School of Pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
| | - Huaqin Yuan
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Qian Li
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Weicheng Wang
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 22 Hankou Road, Nanjing 210093, China.
| | - Xiaomin Li
- Department of Emergency, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China.
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China.
| |
Collapse
|