1
|
Wu H, Mao Y, Wang Q, Yu H, Bouaziz M, Makrides N, Koleske AJ, Radice GL, Zhang X. Abl kinases regulate FGF signaling independent of Crk phosphorylation to prevent Peters anomaly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.619064. [PMID: 39484567 PMCID: PMC11526961 DOI: 10.1101/2024.10.24.619064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Peters anomaly, the most common cause of congenital corneal opacity, stems from corneal-lenticular adhesion. Despite numerous identified mutations, a cohesive molecular framework of the disease's etiology remains elusive. Here, we identified Abl kinases as pivotal regulators of FGF signaling, as genetic ablation of Abl kinases restores lens induction even in the absence of FGF signaling. Intriguingly, both Abl kinase deficiency and increased FGF-Ras activity result in Peters anomaly independent of ERK signaling, which can be rescued by allelic deletion of Abl substrate, Crk. However, contrary to the prevailing belief that Abl kinases regulate Crk proteins by direct phosphorylation, mutations at Abl kinase phosphorylation sites on Crk and CrkL did not yield any observable effects. Instead, our findings reveal that Abl kinases phosphorylate Ptpn12, which in turn inhibits p130Cas phosphorylation and Crk recruitment, crucial for Rho GTPases activation and cytoskeletal dynamics. Consequently, Abl kinase deficiency reduces actomyosin contractility within the lens vesicle and genetically interacts with RhoA inhibition. Conversely, Rac1 deletion mitigates Peters anomaly in models with aberrant FGF, Abl kinase and RhoA signaling. Our results demonstrate that Abl kinases regulate FGF signaling to balance RhoA and Rac1 activity via the Ptpn12-p130Cas pathway, suggesting that targeting tension-mediated lens vesicle separation could be a therapeutic strategy for Peters anomaly.
Collapse
|
2
|
Ma Q, He X, Wang X, Zhao G, Zhang Y, Su C, Wei M, Zhang K, Liu M, Zhu Y, He J. PTPN14 aggravates neointimal hyperplasia via boosting PDGFRβ signaling in smooth muscle cells. Nat Commun 2024; 15:7398. [PMID: 39191789 PMCID: PMC11350182 DOI: 10.1038/s41467-024-51881-x] [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: 11/10/2023] [Accepted: 08/20/2024] [Indexed: 08/29/2024] Open
Abstract
Smooth muscle cell (SMC) phenotypic modulation, primarily driven by PDGFRβ signaling, is implicated in occlusive cardiovascular diseases. However, the promotive and restrictive regulation mechanism of PDGFRβ and the role of protein tyrosine phosphatase non-receptor type 14 (PTPN14) in neointimal hyperplasia remain unclear. Our study observes a marked upregulation of PTPN14 in SMCs during neointimal hyperplasia. PTPN14 overexpression exacerbates neointimal hyperplasia in a phosphatase activity-dependent manner, while SMC-specific deficiency of PTPN14 mitigates this process in mice. RNA-seq indicates that PTPN14 deficiency inhibits PDGFRβ signaling-induced SMC phenotypic modulation. Moreover, PTPN14 interacts with intracellular region of PDGFRβ and mediates its dephosphorylation on Y692 site. Phosphorylation of PDGFRβY692 negatively regulates PDGFRβ signaling activation. The levels of both PTPN14 and phospho-PDGFRβY692 are correlated with the degree of stenosis in human coronary arteries. Our findings suggest that PTPN14 serves as a critical modulator of SMCs, promoting neointimal hyperplasia. PDGFRβY692, dephosphorylated by PTPN14, acts as a self-inhibitory site for controlling PDGFRβ activation.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Coronary Vessels/pathology
- Coronary Vessels/metabolism
- Hyperplasia/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/metabolism
- Neointima/pathology
- Phosphorylation
- Protein Tyrosine Phosphatases, Non-Receptor/metabolism
- Protein Tyrosine Phosphatases, Non-Receptor/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Signal Transduction
Collapse
Affiliation(s)
- Qiannan Ma
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China
- Department of Endocrinology and Metabolism, Tianjin Research Institute of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xue He
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China
| | - Xue Wang
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China
| | - Guobing Zhao
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China
| | - Yanhong Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China
| | - Chao Su
- Division of Cardiovascular Surgery, Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518040, China
| | - Minxin Wei
- Division of Cardiovascular Surgery, Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518040, China
| | - Kai Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Research Institute of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China.
- Department of Endocrinology and Metabolism, Tianjin Research Institute of Endocrinology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Jinlong He
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
3
|
Li W, Huang M, Wu Z, Zhang Y, Cai Y, Su J, Xia J, Yang F, Xiao D, Yang W, Xu Y, Liu Z. mRNA-Lipid Nanoparticle-Mediated Restoration of PTPN14 Exhibits Antitumor Effects by Overcoming Anoikis Resistance in Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309988. [PMID: 39189475 PMCID: PMC11348215 DOI: 10.1002/advs.202309988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/11/2024] [Indexed: 08/28/2024]
Abstract
Triple-negative breast cancer (TNBC) poses a challenging prognosis due to early metastasis driven by anoikis resistance. Identifying crucial regulators to overcome this resistance is vital for improving patient outcomes. In this study, a genome-wide CRISPR/Cas9 knockout screen in TNBC cells has identified tyrosine-protein phosphatase nonreceptor type 14 (PTPN14) as a key regulator of anoikis resistance. PTPN14 expression has shown a progressive decrease from normal breast tissue to metastatic tumors. Overexpressing PTPN14 has induced anoikis and inhibited cell proliferation in TNBC cells, while normal human breast cells are unaffected. Mechanistically, PTPN14 is identified as a key factor in dephosphorylating breast cancer antiestrogen resistance 3, a novel substrate, leading to the subsequent inhibition of PI3K/AKT and ERK signaling pathways. Local delivery of in vitro transcribed PTPN14 mRNA encapsulated by lipid nanoparticles in a TNBC mouse model has effectively inhibited tumor growth and metastasis, prolonging survival. The study underscores PTPN14 as a potential therapeutic target for metastatic TNBC, with the therapeutic strategy based on mRNA expression of PTPN14 demonstrating clinical application prospects in alleviating the burden of both primary tumors and metastatic disease.
Collapse
Affiliation(s)
- Wei Li
- Department of Clinical PharmacologyHunan Key Laboratory of Pharmacogeneticsand National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008P. R. China
- Institute of Clinical PharmacologyEngineering Research Center for applied Technology of Pharmacogenomics of Ministry of EducationCentral South UniversityChangsha410078P. R. China
| | - Masha Huang
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Zhaoping Wu
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangsha410008P. R. China
| | - Yu Zhang
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Ying Cai
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Juncheng Su
- Department of Gastrointestinal SurgeryRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Jia Xia
- Department of NephrologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Fan Yang
- Department of PhysiologySchool of Basic Medical SciencesShandong UniversityJinan250011P. R. China
| | - Desheng Xiao
- Department of PathologySchool of Basic MedicineXiangya HospitalCentral South UniversityChangsha410013P. R. China
| | - Wen Yang
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Zhaoqian Liu
- Department of Clinical PharmacologyHunan Key Laboratory of Pharmacogeneticsand National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008P. R. China
- Institute of Clinical PharmacologyEngineering Research Center for applied Technology of Pharmacogenomics of Ministry of EducationCentral South UniversityChangsha410078P. R. China
| |
Collapse
|
4
|
DeMarco AG, Hall MC. Phosphoproteomic Approaches for Identifying Phosphatase and Kinase Substrates. Molecules 2023; 28:3675. [PMID: 37175085 PMCID: PMC10180314 DOI: 10.3390/molecules28093675] [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: 04/07/2023] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
Protein phosphorylation is a ubiquitous post-translational modification controlled by the opposing activities of protein kinases and phosphatases, which regulate diverse biological processes in all kingdoms of life. One of the key challenges to a complete understanding of phosphoregulatory networks is the unambiguous identification of kinase and phosphatase substrates. Liquid chromatography-coupled mass spectrometry (LC-MS/MS) and associated phosphoproteomic tools enable global surveys of phosphoproteome changes in response to signaling events or perturbation of phosphoregulatory network components. Despite the power of LC-MS/MS, it is still challenging to directly link kinases and phosphatases to specific substrate phosphorylation sites in many experiments. Here, we survey common LC-MS/MS-based phosphoproteomic workflows for identifying protein kinase and phosphatase substrates, noting key advantages and limitations of each. We conclude by discussing the value of inducible degradation technologies coupled with phosphoproteomics as a new approach that overcomes some limitations of current methods for substrate identification of kinases, phosphatases, and other regulatory enzymes.
Collapse
Affiliation(s)
- Andrew G. DeMarco
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Mark C. Hall
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
5
|
Li H, Guan B, Liu S, Liu H, Song L, Zhang G, Zhao R, Zhou C, Gao P. PTPN14 promotes gastric cancer progression by PI3KA/AKT/mTOR pathway. Cell Death Dis 2023; 14:188. [PMID: 36898991 PMCID: PMC10006225 DOI: 10.1038/s41419-023-05712-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/12/2023]
Abstract
Gastric cancer is a high molecular heterogeneous disease with a poor prognosis. Although gastric cancer is a hot area of medical research, the mechanism of gastric cancer occurrence and development is still unclear. New strategies for treating gastric cancer need to be further explored. Protein tyrosine phosphatases play vital roles in cancer. A growing stream of studies shows that strategies or inhibitors targeting protein tyrosine phosphatases have been developed. PTPN14 belongs to the protein tyrosine phosphatase subfamily. As an inert phosphatase, PTPN14 has very poor activity and mainly functions as a binding protein through its FERM (four-point-one, ezrin, radixin, and moesin) domain or PPxY motif. The online database indicated that PTPN14 may be a poor prognostic factor for gastric cancer. However, the function and underlying mechanism of PTPN14 in gastric cancer remain unclear. We collected gastric cancer tissues and detected the expression of PTPN14. We found that PTPN14 was elevated in gastric cancer. Further correlation analysis indicated that PTPN14 was relevant with the T stage and cTNM (clinical tumor node metastasis classification) stage. The survival curve analysis showed that gastric cancer patients with higher PTPN14 expression had a shorter survival time. In addition, we illustrated that CEBP/β (CCAAT enhanced binding protein beta) could transcriptionally activate PTPN14 expression in gastric cancer. The highly expressed PTPN14 combined with NFkB (nuclear factor Kappa B) through its FERM domain and accelerated NFkB nucleus translocation. Then, NFkB promoted the transcription of PI3KA and initiated the PI3KA/AKT/mTOR pathway to promote gastric cancer cell proliferation, migration, and invasion. Finally, we established mice models to validate the function and the molecular mechanism of PTPN14 in gastric cancer. In summary, our results illustrated the function of PTPN14 in gastric cancer and demonstrated the potential mechanisms. Our findings provide a theoretical basis to better understand the occurrence and development of gastric cancer.
Collapse
Affiliation(s)
- Hui Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Shandong University, 250012, Jinan, Shandong, China
- Department of Pathology, Qilu Hospital, Shandong University, 250012, Jinan, Shandong, China
- Department of Pathology, The Second Hospital of Shandong University, 250012, Jinan, Shandong, China
| | - Bingxin Guan
- Department of Pathology, The Second Hospital of Shandong University, 250012, Jinan, Shandong, China
| | - Sen Liu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Shandong University, 250012, Jinan, Shandong, China
- Department of Pathology, Qilu Hospital, Shandong University, 250012, Jinan, Shandong, China
| | - Haiting Liu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Shandong University, 250012, Jinan, Shandong, China
- Department of Pathology, Qilu Hospital, Shandong University, 250012, Jinan, Shandong, China
| | - Lin Song
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Shandong University, 250012, Jinan, Shandong, China
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Guohao Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Shandong University, 250012, Jinan, Shandong, China
- Department of Pathology, Qilu Hospital, Shandong University, 250012, Jinan, Shandong, China
| | - Ruinan Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Shandong University, 250012, Jinan, Shandong, China
- Department of Pathology, Qilu Hospital, Shandong University, 250012, Jinan, Shandong, China
| | - Chengjun Zhou
- Department of Pathology, The Second Hospital of Shandong University, 250012, Jinan, Shandong, China
| | - Peng Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Shandong University, 250012, Jinan, Shandong, China.
- Department of Pathology, Qilu Hospital, Shandong University, 250012, Jinan, Shandong, China.
| |
Collapse
|
6
|
Emerging Role of Plant-Based Dietary Components in Post-Translational Modifications Associated with Colorectal Cancer. Life (Basel) 2023; 13:life13020264. [PMID: 36836621 PMCID: PMC9962725 DOI: 10.3390/life13020264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide. Its main modifiable risk factors are diet, alcohol consumption, and smoking. Thus, the right approach through lifestyle changes may lead to its prevention. In fact, some natural dietary components have exhibited chemopreventive activity through modulation of cellular processes involved in CRC development. Although cancer is a multi-factorial process, the study of post-translational modifications (PTMs) of proteins associated with CRC has recently gained interest, as inappropriate modification is closely related to the activation of cell signalling pathways involved in carcinogenesis. Therefore, this review aimed to collect the main PTMs associated with CRC, analyse the relationship between different proteins that are susceptible to inappropriate PTMs, and review the available scientific literature on the role of plant-based dietary compounds in modulating CRC-associated PTMs. In summary, this review suggested that some plant-based dietary components such as phenols, flavonoids, lignans, terpenoids, and alkaloids may be able to correct the inappropriate PTMs associated with CRC and promote apoptosis in tumour cells.
Collapse
|
7
|
The tyrosine phosphatase PTPN14 inhibits the activation of STAT3 in PEDV infected Vero cells. Vet Microbiol 2022; 267:109391. [DOI: 10.1016/j.vetmic.2022.109391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 11/23/2022]
|
8
|
Zhang D, Wu F, Song J, Meng M, Fan X, Lu C, Weng Q, Fang S, Zheng L, Tang B, Yang Y, Tu J, Xu M, Zhao Z, Ji J. A role for the NPM1/PTPN14/YAP axis in mediating hypoxia-induced chemoresistance to sorafenib in hepatocellular carcinoma. Cancer Cell Int 2022; 22:65. [PMID: 35135548 PMCID: PMC8822852 DOI: 10.1186/s12935-022-02479-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/20/2022] [Indexed: 12/21/2022] Open
Abstract
Background Tumor microenvironments are characterized by resistance to chemotherapeutic agents and radiotherapy. Hypoxia plays an important role in the development of tumor resistance, as well as the generation of metastatic potential. YAP also participates in the regulation of hypoxia-mediated chemoresistance, and is negatively regulated by protein tyrosine phosphatase non-receptor type 14 (PTPN14). Methods The PTPN14 expression in hepatocellular carcinoma (HCC) tissues were evaluated by qRT-PCR, western blot and tissue microarrays. The effect of PTPN14 on HCC progression was investigated in vitro and in vivo. Results Here, we report that PTPN14 expression was downregulated in HCC tissues and cell lines. Silencing PTPN14 significantly enhanced proliferation, migration, invasion of HepG2 cells in vitro and tumor growth and metastasis in vivo, whereas overexpression of PTPN14 significantly inhibited these abilities in SK-Hep1 cells. We also found that hypoxia-induced nuclear translocation and accumulation of PTPN14 led to resistance to sorafenib in HCC cells. Further mechanistic studies suggested that NPM1 regulates PTPN14 localization, and that NPM1 regulates YAP by retaining PTPN14 in the nucleus under hypoxic conditions. Conclusions These data suggest that a therapeutic strategy against chemoresistant HCC may involve disruption of NPM1-mediated regulation of YAP by retaining PTPN14 in the nucleus under hypoxic conditions. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02479-0.
Collapse
Affiliation(s)
- Dengke Zhang
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Fazong Wu
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Jingjing Song
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Miaomiao Meng
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Xiaoxi Fan
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Chenying Lu
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Qiaoyou Weng
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Shiji Fang
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Liyun Zheng
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Bufu Tang
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Yang Yang
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Jianfei Tu
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Min Xu
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Jiansong Ji
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| |
Collapse
|
9
|
Kumbrink J, Li P, Pók-Udvari A, Klauschen F, Kirchner T, Jung A. p130Cas Is Correlated with EREG Expression and a Prognostic Factor Depending on Colorectal Cancer Stage and Localization Reducing FOLFIRI Efficacy. Int J Mol Sci 2021; 22:ijms222212364. [PMID: 34830244 PMCID: PMC8625396 DOI: 10.3390/ijms222212364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/31/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022] Open
Abstract
p130 Crk-associated substrate (p130Cas) is associated with poor prognosis and treatment resistance in breast and lung cancers. To elucidate p130Cas functional and clinical role in colorectal cancer (CRC) progression/therapy resistance, we performed cell culture experiments and bioinformatic/statistical analyses of clinical data sets. p130Cas expression was associated with poor survival in the cancer genome atlas (TCGA) data set. Knockdown/reconstitution experiments showed that p130Cas drives migration but, unexpectedly, inhibits proliferation in CRC cells. TCGA data analyses identified the growth factor epiregulin (EREG) as inversely correlated with p130Cas. p130Cas knockdown and simultaneous EREG treatment further enhanced proliferation. RNA interference and EREG treatment experiments suggested that p130Cas/EREG limit each other’s expression/activity. Inverse p130Cas/EREG Spearman correlations were prominent in right-sided and earlier stage CRC. p130Cas was inducible by 5-fluorouracil (5-FU) and FOLFIRI (folinic acid, 5-FU, irinotecan), and p130Cas and EREG were upregulated in distant metastases (GSE121418). Positive p130Cas/EREG correlations were observed in metastases, preferentially in post-treatment samples (especially pulmonary metastases). p130Cas knockdown sensitized CRC cells to FOLFIRI independent of EREG treatment. RNA sequencing and gene ontology analyses revealed that p130Cas is involved in cytochrome P450 drug metabolism and epithelial-mesenchymal transition. p130Cas expression was associated with poor survival in right-sided, stage I/II, MSS (microsatellite stable), or BRAF-mutated CRC. In summary, p130Cas represents a prognostic factor and potential therapeutic target in CRC.
Collapse
Affiliation(s)
- Jörg Kumbrink
- Faculty of Medicine, Institute of Pathology, Ludwig-Maximilians-University of Munich, 80337 Munich, Germany; (P.L.); (A.P.-U.); (F.K.); (T.K.); (A.J.)
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
- Correspondence:
| | - Pan Li
- Faculty of Medicine, Institute of Pathology, Ludwig-Maximilians-University of Munich, 80337 Munich, Germany; (P.L.); (A.P.-U.); (F.K.); (T.K.); (A.J.)
| | - Agnes Pók-Udvari
- Faculty of Medicine, Institute of Pathology, Ludwig-Maximilians-University of Munich, 80337 Munich, Germany; (P.L.); (A.P.-U.); (F.K.); (T.K.); (A.J.)
| | - Frederick Klauschen
- Faculty of Medicine, Institute of Pathology, Ludwig-Maximilians-University of Munich, 80337 Munich, Germany; (P.L.); (A.P.-U.); (F.K.); (T.K.); (A.J.)
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Thomas Kirchner
- Faculty of Medicine, Institute of Pathology, Ludwig-Maximilians-University of Munich, 80337 Munich, Germany; (P.L.); (A.P.-U.); (F.K.); (T.K.); (A.J.)
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Andreas Jung
- Faculty of Medicine, Institute of Pathology, Ludwig-Maximilians-University of Munich, 80337 Munich, Germany; (P.L.); (A.P.-U.); (F.K.); (T.K.); (A.J.)
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| |
Collapse
|
10
|
Nayak S, Bhatt MLB, Goel MM, Gupta S, Mehrotra D, Mahdi AA, Mishra A. Aberrant Expression of PTPN-14 and Wilms’ Tumor 1 as Putative Biomarker for Locoregional Recurrence in Oral Squamous Cell Carcinoma. ASIAN JOURNAL OF ONCOLOGY 2021. [DOI: 10.1055/s-0041-1731128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Abstract
Objective Locoregional recurrence in oral squamous cell carcinoma (OSCC) is a major concern that leads to metastasis. Its detection at earliest stage is very important to increase the overall survival of the patient. There is no any biomarker for locoregional recurrence in oral squamous cell carcinoma (OSCC). The aim of this study was to find a biomarker for locoregional recurrence in tissue and serum at gene and protein level.
Methods This work studied the expression of protein tyrosine phosphatase nonreceptor type 14 (PTPN-14) and Wilms’ tumor 1 (WT-1) in patients and correlated their expression with locoregional recurrence and survival. Tissue expression was observed in formalin fixed tissue biopsies of 96 OSCC and 32 healthy controls by immunohistochemistry using antibody against PTPN-14 and WT-1 and serum level was estimated by enzyme-linked immunosorbent assay in pre- and post-chemoradiotherapy samples. mRNA expression was determined by using real-time polymerase chain reaction. Patients were followed for 3 years for locoregional recurrence.
Results Expression of PTPN-14 and WT-1 in OSCC was upregulated (aberrant) in tissue and sera in both gene and protein level as compared with healthy controls. Locoregional recurrence was observed in 10 (23.80%) patients and significantly associated with PTPN-14 (p < 0.047) and WT-1 expression (p < 0.031).
Conclusion PTPN-14 and WT-1 may be used as biomarker to identify patients for higher risk of locoregional recurrence. This study drove molecular aspect and phenotypic level to derive new emergent strategies in future for recurrent OSCC.
Collapse
Affiliation(s)
- Seema Nayak
- Department of Radiotherapy, King George’s Medical University, Lucknow, Uttar Pradesh, India
- Medanta Holding Pvt. Ltd., Lucknow, Uttar Pradesh, India
| | - Madan Lal Brahma Bhatt
- Department of Radiotherapy, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Madhu Mati Goel
- Medanta Holding Pvt. Ltd., Lucknow, Uttar Pradesh, India
- Department of Pathology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Seema Gupta
- Department of Radiotherapy, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Divya Mehrotra
- Department of Oral and Maxillofacial Surgery, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Abbas Ali Mahdi
- Department of Biochemistry, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Anupam Mishra
- Department of Otorhinolaryngology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
11
|
Monoe Y, Jingushi K, Kawase A, Hirono T, Hirose R, Nakatsuji Y, Kitae K, Ueda Y, Hase H, Abe Y, Adachi J, Tomonaga T, Tsujikawa K. Pharmacological Inhibition of miR-130 Family Suppresses Bladder Tumor Growth by Targeting Various Oncogenic Pathways via PTPN1. Int J Mol Sci 2021; 22:ijms22094751. [PMID: 33947152 PMCID: PMC8124864 DOI: 10.3390/ijms22094751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/24/2021] [Accepted: 04/28/2021] [Indexed: 12/25/2022] Open
Abstract
Previously, we have revealed that the miR-130 family (miR-130b, miR-301a, and miR-301b) functions as an oncomiR in bladder cancer. The pharmacological inhibition of the miR-130 family molecules by the seed-targeting strategy with an 8-mer tiny locked nucleic acid (LNA) inhibits the growth, migration, and invasion of bladder cancer cells by repressing stress fiber formation. Here, we searched for a functionally advanced target sequence with LNA for the miR-130 family with low cytotoxicity and found LNA #9 (A(L)^i^i^A(L)^T(L)^T(L)^G(L)^5(L)^A(L)^5(L)^T(L)^G) as a candidate LNA. LNA #9 inhibited cell growth in vitro and in an in vivo orthotopic bladder cancer model. Proteome-wide tyrosine phosphorylation analysis suggested that the miR-130 family upregulates a wide range of receptor tyrosine kinases (RTKs) signaling via the expression of phosphorylated Src (pSrcTyr416). SILAC-based proteome analysis and a luciferase assay identified protein tyrosine phosphatase non-receptor type 1 (PTPN1), which is implicated as a negative regulator of multiple signaling pathways downstream of RTKs as a target gene of the miR-130 family. The miR-130-targeted LNA increased and decreased PTPN1 and pSrcTyr416 expressions, respectively. PTPN1 knockdown led to increased tumor properties (cell growth, invasion, and migration) and increased pSrcTyr416 expression in bladder cancer cells, suggesting that the miR-130 family upregulates multiple RTK signaling by targeting PTPN1 and subsequent Src activation in bladder cancer. Thus, our newly designed miR-130 family targeting LNA could be a promising nucleic acid therapeutic agent for bladder cancer.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Carcinoma, Transitional Cell/drug therapy
- Carcinoma, Transitional Cell/genetics
- Carcinoma, Transitional Cell/metabolism
- Cell Line, Tumor
- Drug Screening Assays, Antitumor
- Female
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Humans
- Mice
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- Neoplasm Proteins/physiology
- Oligonucleotides/therapeutic use
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology
- RNA, Neoplasm/antagonists & inhibitors
- RNA, Neoplasm/genetics
- Receptor Protein-Tyrosine Kinases/biosynthesis
- Receptor Protein-Tyrosine Kinases/genetics
- Recombinant Proteins/metabolism
- Up-Regulation
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yuya Monoe
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.M.); (A.K.); (T.H.); (R.H.); (Y.N.); (K.K.); (Y.U.); (H.H.); (K.T.)
| | - Kentaro Jingushi
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.M.); (A.K.); (T.H.); (R.H.); (Y.N.); (K.K.); (Y.U.); (H.H.); (K.T.)
- Correspondence: ; Tel.: +81-6-6879-8192
| | - Akitaka Kawase
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.M.); (A.K.); (T.H.); (R.H.); (Y.N.); (K.K.); (Y.U.); (H.H.); (K.T.)
| | - Takayuki Hirono
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.M.); (A.K.); (T.H.); (R.H.); (Y.N.); (K.K.); (Y.U.); (H.H.); (K.T.)
| | - Ryo Hirose
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.M.); (A.K.); (T.H.); (R.H.); (Y.N.); (K.K.); (Y.U.); (H.H.); (K.T.)
| | - Yoshino Nakatsuji
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.M.); (A.K.); (T.H.); (R.H.); (Y.N.); (K.K.); (Y.U.); (H.H.); (K.T.)
| | - Kaori Kitae
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.M.); (A.K.); (T.H.); (R.H.); (Y.N.); (K.K.); (Y.U.); (H.H.); (K.T.)
| | - Yuko Ueda
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.M.); (A.K.); (T.H.); (R.H.); (Y.N.); (K.K.); (Y.U.); (H.H.); (K.T.)
| | - Hiroaki Hase
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.M.); (A.K.); (T.H.); (R.H.); (Y.N.); (K.K.); (Y.U.); (H.H.); (K.T.)
| | - Yuichi Abe
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan; (Y.A.); (J.A.); (T.T.)
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Jun Adachi
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan; (Y.A.); (J.A.); (T.T.)
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan; (Y.A.); (J.A.); (T.T.)
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.M.); (A.K.); (T.H.); (R.H.); (Y.N.); (K.K.); (Y.U.); (H.H.); (K.T.)
| |
Collapse
|
12
|
Wang Q, Yu C, Yue C, Liu X. Fusobacterium nucleatum produces cancer stem cell characteristics via EMT-resembling variations. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:1819-1828. [PMID: 32782710 PMCID: PMC7414483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/28/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To explore the involvement of epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) characteristics induced by Fusobacterium nucleatum (Fn) in colorectal cancer (CRC) in vitro. METHODS SW480 and HCT116 cells were co-cultivated with Fn. Western blot (WB) and real-time PCR were used for detecting EMT markers' expression. CSC-resembling phenotypes were observed through migration, intrusion, and spherical colony formation assays. Flow cytometry was employed for sorting, based on the expression of CD44. RESULTS It was displayed that Fn infection was responsible for an EMT phenotype associated with an increase in mesenchymal markers (Snail1, Vimentin, and ZEB1) as well as CD44 expression. Fn treatment induced stronger expressions of such markers when MOI increased. Furthermore, infection resulted in augmented migration, intrusion, and tumorsphere formation capacities. Cell classification implicated that mere CD44high cells exhibited CSC characteristics and mesenchymal phenotype (MP) in vitro, accompanied with augmented tumor-causing capacity over CD44low cells. Finally, we demonstrated IL-6/STAT3 pathway was involved in EMT-CSC-resembling behavior of CRC cells. CONCLUSION All of these data suggest that Fn reveals CSC-resembling characteristics through activating IL-6/STAT3 and eliciting EMT-resembling variations in colorectal epithelial cells (CECs).
Collapse
Affiliation(s)
- Qin Wang
- Department of Clinical Medicine, Jiangsu Health Vocational CollegeNanjing, China
| | - Chen Yu
- Department of Integrated Traditional Chinese and Western Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical UniversityNanjing, China
| | - Chao Yue
- Department of General Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical UniversityNanjing, China
| | - Xin Liu
- Department of Prevention and Control for Occupational Disease, Jiangsu Provincial Center for Disease Prevention and ControlNanjing, China
| |
Collapse
|
13
|
Reiterer V, Pawłowski K, Desrochers G, Pause A, Sharpe HJ, Farhan H. The dead phosphatases society: a review of the emerging roles of pseudophosphatases. FEBS J 2020; 287:4198-4220. [PMID: 32484316 DOI: 10.1111/febs.15431] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/12/2020] [Accepted: 05/27/2020] [Indexed: 12/11/2022]
Abstract
Phosphatases are a diverse family of enzymes, comprising at least 10 distinct protein folds. Like most other enzyme families, many have sequence variations that predict an impairment or loss of catalytic activity classifying them as pseudophosphatases. Research on pseudoenzymes is an emerging area of interest, with new biological functions repurposed from catalytically active relatives. Here, we provide an overview of the pseudophosphatases identified to date in all major phosphatase families. We will highlight the degeneration of the various catalytic sequence motifs and discuss the challenges associated with the experimental determination of catalytic inactivity. We will also summarize the role of pseudophosphatases in various diseases and discuss the major challenges and future directions in this field.
Collapse
Affiliation(s)
| | | | - Guillaume Desrochers
- Department of Biochemistry, McGill University, Montréal, QC, Canada.,Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
| | - Arnim Pause
- Department of Biochemistry, McGill University, Montréal, QC, Canada.,Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
| | | | - Hesso Farhan
- Institute of Basic Medical Sciences, University of Oslo, Norway
| |
Collapse
|
14
|
Chen J, Zhao X, Yuan Y, Jing JJ. The expression patterns and the diagnostic/prognostic roles of PTPN family members in digestive tract cancers. Cancer Cell Int 2020; 20:238. [PMID: 32536826 PMCID: PMC7291430 DOI: 10.1186/s12935-020-01315-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background Non-receptor protein tyrosine phosphatases (PTPNs) are a set of enzymes involved in the tyrosyl phosphorylation. The present study intended to clarify the associations between the expression patterns of PTPN family members, and diagnosis as well as the prognosis of digestive tract cancers. Methods Oncomine and Ualcan were used to analyze PTPN expressions. Data from The Cancer Genome Atlas (TCGA) were downloaded through UCSC Xena for validation and to explore the relationship of the PTPN expression with diagnosis, clinicopathological parameters and survival of digestive tract cancers. Gene ontology enrichment analysis was conducted using the DAVID database. The gene–gene interaction network was performed by GeneMANIA and the protein–protein interaction (PPI) network was built using STRING portal coupled with Cytoscape. The expression of differentially expressed PTPNs in cancer cell lines were explored using CCLE. Moreover, by histological verification, the expression of four PTPNs in digestive tract cancers were further analyzed. Results Most PTPN family members were associated with digestive tract cancers according to Oncomine, Ualcan and TCGA data. Several PTPN members were differentially expressed in digestive tract cancers. For esophageal carcinoma (ESCA), PTPN1 and PTPN12 levels were correlated with incidence; PTPN20 was associated with poor prognosis. For stomach adenocarcinoma (STAD), PTPN2 and PTPN12 levels were correlated with incidence; PTPN3, PTPN5, PTPN7, PTPN11, PTPN13, PTPN14, PTPN18 and PTPN23 were correlated with pathological grade; PTPN20 expression was related with both TNM stage and N stage; PTPN22 was associated with T stage and pathological grade; decreased expression of PTPN5 and PTPN13 implied worse overall survival of STAD, while elevated PTPN6 expression indicated better prognosis. For colorectal cancer (CRC), PTPN2, PTPN21 and PTPN22 levels were correlated with incidence; expression of PTPN5, PTPN12, and PTPN14 was correlated with TNM stage and N stage; high PTPN5 or PTPN7 expression was associated with increased hazards of death. CCLE analyses showed that in esophagus cancer cell lines, PTPN1, PTPN4 and PTPN12 were highly expressed; in gastric cancer cell lines, PTPN2 and PTPN12 were highly expressed; in colorectal cancer cell lines, PTPN12 was highly expressed while PTPN22 was downregulated. Results of histological verification experiment showed differential expressions of PTPN22 in CRC, and PTPN12 in GC and CRC. Conclusions Members of PTPN family were differentially expressed in digestive tract cancers. Correlations were found between PTPN genes and clinicopathological parameters of patients. Expression of PTPN12 was upregulated in both STAD and CRC, and thus could be used as a diagnostic biomarker. Differential expression of PTPN12 in GC and CRC, and PTPN22 in CRC were presented in our histological verification experiment.
Collapse
Affiliation(s)
- Jing Chen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning People's Republic of China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, 110001 China.,Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001 China
| | - Xu Zhao
- Mathematical Computer Teaching and Research Office, Liaoning Vocational College of Medicine, Shenyang, 110101 China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning People's Republic of China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, 110001 China.,Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001 China
| | - Jing-Jing Jing
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning People's Republic of China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, 110001 China.,Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, 110001 China
| |
Collapse
|
15
|
Díaz-Valdivia NI, Díaz J, Contreras P, Campos A, Rojas-Celis V, Burgos-Ravanal RA, Lobos-González L, Torres VA, Perez VI, Frei B, Leyton L, Quest AFG. The non-receptor tyrosine phosphatase type 14 blocks caveolin-1-enhanced cancer cell metastasis. Oncogene 2020; 39:3693-3709. [PMID: 32152405 PMCID: PMC7190567 DOI: 10.1038/s41388-020-1242-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 01/20/2020] [Accepted: 02/21/2020] [Indexed: 01/13/2023]
Abstract
Caveolin-1 (CAV1) enhanced migration, invasion, and metastasis of cancer cells is inhibited by co-expression of the glycoprotein E-cadherin. Although the two proteins form a multiprotein complex that includes β-catenin, it remained unclear how this would contribute to blocking the metastasis promoting function of CAV1. Here, we characterized by mass spectrometry the protein composition of CAV1 immunoprecipitates from B16F10 murine melanoma cells expressing or not E-cadherin. The novel protein tyrosine phosphatase PTPN14 was identified by mass spectrometry analysis exclusively in co-immunoprecipitates of CAV1 with E-cadherin. Interestingly, PTPN14 is implicated in controlling metastasis, but only few known PTPN14 substrates exist. We corroborated by western blotting experiments that PTPN14 and CAV1 co-inmunoprecipitated in the presence of E-cadherin in B16F10 melanoma and other cancer cells. Moreover, the CAV1(Y14F) mutant protein was shown to co-immunoprecipitate with PTPN14 even in the absence of E-cadherin, and overexpression of PTPN14 reduced CAV1 phosphorylation on tyrosine-14, as well as suppressed CAV1-enhanced cell migration, invasion and Rac-1 activation in B16F10, metastatic colon [HT29(US)] and breast cancer (MDA-MB-231) cell lines. Finally, PTPN14 overexpression in B16F10 cells reduced the ability of CAV1 to induce metastasis in vivo. In summary, we identify here CAV1 as a novel substrate for PTPN14 and show that overexpression of this phosphatase suffices to reduce CAV1-induced metastasis.
Collapse
Affiliation(s)
- Natalia I Díaz-Valdivia
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Jorge Díaz
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Institute for Research in Dental Science, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Pamela Contreras
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - América Campos
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Fundación Ciencia & Vida, Santiago, Chile
| | - Victoria Rojas-Celis
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Renato A Burgos-Ravanal
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lorena Lobos-González
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Fundación Ciencia & Vida, Santiago, Chile
| | - Vicente A Torres
- Institute for Research in Dental Science, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Viviana I Perez
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Balz Frei
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Lisette Leyton
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| | - Andrew F G Quest
- Cellular Communication Laboratory, Center for studies on Exercise, Metabolism and Cancer (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
16
|
PTPN14 degradation by high-risk human papillomavirus E7 limits keratinocyte differentiation and contributes to HPV-mediated oncogenesis. Proc Natl Acad Sci U S A 2019; 116:7033-7042. [PMID: 30894485 DOI: 10.1073/pnas.1819534116] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
High-risk human papillomavirus (HPV) E7 proteins enable oncogenic transformation of HPV-infected cells by inactivating host cellular proteins. High-risk but not low-risk HPV E7 target PTPN14 for proteolytic degradation, suggesting that PTPN14 degradation may be related to their oncogenic activity. HPV infects human keratinocytes but the role of PTPN14 in keratinocytes and the consequences of PTPN14 degradation are unknown. Using an HPV16 E7 variant that can inactivate retinoblastoma tumor suppressor (RB1) but cannot degrade PTPN14, we found that high-risk HPV E7-mediated PTPN14 degradation impairs keratinocyte differentiation. Deletion of PTPN14 from primary human keratinocytes decreased keratinocyte differentiation gene expression. Related to oncogenic transformation, both HPV16 E7-mediated PTPN14 degradation and PTPN14 deletion promoted keratinocyte survival following detachment from a substrate. PTPN14 degradation contributed to high-risk HPV E6/E7-mediated immortalization of primary keratinocytes and HPV+ but not HPV- cancers exhibit a gene-expression signature consistent with PTPN14 inactivation. We find that PTPN14 degradation impairs keratinocyte differentiation and propose that this contributes to high-risk HPV E7-mediated oncogenic activity independent of RB1 inactivation.
Collapse
|
17
|
Bottini A, Wu DJ, Ai R, Le Roux M, Bartok B, Bombardieri M, Doody KM, Zhang V, Sacchetti C, Zoccheddu M, Lonic A, Li X, Boyle DL, Hammaker D, Meng TC, Liu L, Corr M, Stanford SM, Lewis M, Wang W, Firestein GS, Khew-Goodall Y, Pitzalis C, Bottini N. PTPN14 phosphatase and YAP promote TGFβ signalling in rheumatoid synoviocytes. Ann Rheum Dis 2019; 78:600-609. [PMID: 30808624 DOI: 10.1136/annrheumdis-2018-213799] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 11/04/2022]
Abstract
OBJECTIVE We aimed to understand the role of the tyrosine phosphatase PTPN14-which in cancer cells modulates the Hippo pathway by retaining YAP in the cytosol-in fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis (RA). METHODS Gene/protein expression levels were measured by quantitative PCR and/or Western blotting. Gene knockdown in RA FLS was achieved using antisense oligonucleotides. The interaction between PTPN14 and YAP was assessed by immunoprecipitation. The cellular localisation of YAP and SMAD3 was examined via immunofluorescence. SMAD reporter studies were carried out in HEK293T cells. The RA FLS/cartilage coimplantation and passive K/BxN models were used to examine the role of YAP in arthritis. RESULTS RA FLS displayed overexpression of PTPN14 when compared with FLS from patients with osteoarthritis (OA). PTPN14 knockdown in RA FLS impaired TGFβ-dependent expression of MMP13 and potentiation of TNF signalling. In RA FLS, PTPN14 formed a complex with YAP. Expression of PTPN14 or nuclear YAP-but not of a non-YAP-interacting PTPN14 mutant-enhanced SMAD reporter activity. YAP promoted TGFβ-dependent SMAD3 nuclear localisation in RA FLS. Differences in epigenetic marks within Hippo pathway genes, including YAP, were found between RA FLS and OA FLS. Inhibition of YAP reduced RA FLS pathogenic behaviour and ameliorated arthritis severity. CONCLUSION In RA FLS, PTPN14 and YAP promote nuclear localisation of SMAD3. YAP enhances a range of RA FLS pathogenic behaviours which, together with epigenetic evidence, points to the Hippo pathway as an important regulator of RA FLS behaviour.
Collapse
Affiliation(s)
- Angel Bottini
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Dennis J Wu
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Rizi Ai
- Dept. of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Michelle Le Roux
- Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Beatrix Bartok
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Karen M Doody
- Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Vida Zhang
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Cristiano Sacchetti
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Martina Zoccheddu
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Ana Lonic
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Xiaochun Li
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - David L Boyle
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Deepa Hammaker
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Tzu-Ching Meng
- Institute for Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Lin Liu
- Dept. of Family Medicine and Public Health, University of California San Diego, La Jolla, CA, USA
| | - Maripat Corr
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Stephanie M Stanford
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Myles Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Wei Wang
- Dept. of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.,Dept. of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gary S Firestein
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Yeesim Khew-Goodall
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nunzio Bottini
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA .,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| |
Collapse
|
18
|
Choi J, Saraf A, Florens L, Washburn MP, Busino L. PTPN14 regulates Roquin2 stability by tyrosine dephosphorylation. Cell Cycle 2018; 17:2243-2255. [PMID: 30209976 DOI: 10.1080/15384101.2018.1522912] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Protein phosphorylation regulates a variety of cellular signaling pathways and fundamental mechanisms in cells. In this paper, we demonstrate that the mRNA decay factor Roquin2 is phosphorylated at tyrosine residue in position 691 in vivo. This phosphorylation disrupts the interaction with KLHL6, the E3 ligase for Roquin2. Furthermore, we establish that the tyrosine phosphatase PTPN14 specifically interacts with Roquin2 through its phosphatase domain and dephosphorylates Roquin2 tyrosine 691. Overexpression of PTPN14 promotes Roquin2 degradation in a KLHL6-dependant manner by promoting interaction with KLHL6. Collectively, our findings reveal that PTPN14 negatively regulates the protein stability of Roquin2, thereby adding a new layer of regulation to the KLHL6-Roquin2 axis.
Collapse
Affiliation(s)
- Jaewoo Choi
- a Department of Cancer Biology, Perelman School of Medicine , University of Pennsylvania , Philadelphia , PA , USA
| | - Anita Saraf
- b The Stowers Institute of Medical Research , Kansas , MO , USA
| | | | - Michael P Washburn
- b The Stowers Institute of Medical Research , Kansas , MO , USA.,c Department of Pathology and Laboratory Medicine , The University of Kansas Medical Center , Kansas , KS , USA
| | - Luca Busino
- a Department of Cancer Biology, Perelman School of Medicine , University of Pennsylvania , Philadelphia , PA , USA
| |
Collapse
|
19
|
Zhao Y, Scott A, Zhang P, Hao Y, Feng X, Somasundaram S, Khalil AM, Willis J, Wang Z. Regulation of paxillin-p130-PI3K-AKT signaling axis by Src and PTPRT impacts colon tumorigenesis. Oncotarget 2018; 8:48782-48793. [PMID: 27447856 PMCID: PMC5564724 DOI: 10.18632/oncotarget.10654] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/03/2016] [Indexed: 01/31/2023] Open
Abstract
Protein tyrosine phosphatase receptor T (PTPRT) is frequently mutated in a variety of human cancers including colorectal cancer. Here we report that PTPRT knockout increases the size of mouse colon tumors in the Apcmin+/− genetic background, suggesting that inactivation of PTPRT promotes tumor progression. We previously demonstrated that PTPRT dephosphorylates paxillin at tyrosine-Y88 residue. Consistently, phosphorylation of Y88 paxillin (pY88) is up-regulated in colon tumors derived from Apcmin+/− Ptprt−/− mice. An important downstream effector of pY88 paxillin is the oncogene Akt. Here, we show that pY88 paxillin impacts the Akt pathway by regulating the interaction between p130cas and the p85 regulatory subunit of PI3-Kinase. Additionally, while pY88 paxillin is a substrate of the tumor suppressor phosphatase PTPRT, the corresponding kinase has not been previously identified. In this study, we demonstrate that the oncogenic kinase Src directly phosphorylates paxillin at Y88. Moreover, colorectal cancer cells that express high levels of pY88 paxillin are sensitive to dasatinib treatment, suggesting that pY88 paxillin may serve as a predictive biomarker for Src family kinase inhibitors.
Collapse
Affiliation(s)
- Yiqing Zhao
- Department of Genetics and Genome Sciences, Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Anthony Scott
- Department of Genetics and Genome Sciences, Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Peng Zhang
- Department of Genetics and Genome Sciences, Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Yujun Hao
- Department of Genetics and Genome Sciences, Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Xiujing Feng
- Department of Genetics and Genome Sciences, Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Saigopal Somasundaram
- Department of Genetics and Genome Sciences, Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Ahmad M Khalil
- Department of Genetics and Genome Sciences, Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Joseph Willis
- Department of Pathology, Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Zhenghe Wang
- Department of Genetics and Genome Sciences, Case Medical Center and Case Western Reserve University, Cleveland, Ohio 44106, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| |
Collapse
|
20
|
Meeusen B, Janssens V. Tumor suppressive protein phosphatases in human cancer: Emerging targets for therapeutic intervention and tumor stratification. Int J Biochem Cell Biol 2017; 96:98-134. [PMID: 29031806 DOI: 10.1016/j.biocel.2017.10.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
Aberrant protein phosphorylation is one of the hallmarks of cancer cells, and in many cases a prerequisite to sustain tumor development and progression. Like protein kinases, protein phosphatases are key regulators of cell signaling. However, their contribution to aberrant signaling in cancer cells is overall less well appreciated, and therefore, their clinical potential remains largely unexploited. In this review, we provide an overview of tumor suppressive protein phosphatases in human cancer. Along their mechanisms of inactivation in defined cancer contexts, we give an overview of their functional roles in diverse signaling pathways that contribute to their tumor suppressive abilities. Finally, we discuss their emerging roles as predictive or prognostic markers, their potential as synthetic lethality targets, and the current feasibility of their reactivation with pharmacologic compounds as promising new cancer therapies. We conclude that their inclusion in clinical practice has obvious potential to significantly improve therapeutic outcome in various ways, and should now definitely be pushed forward.
Collapse
Affiliation(s)
- Bob Meeusen
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium.
| |
Collapse
|
21
|
Mello SS, Valente LJ, Raj N, Seoane JA, Flowers BM, McClendon J, Bieging-Rolett KT, Lee J, Ivanochko D, Kozak MM, Chang DT, Longacre TA, Koong AC, Arrowsmith CH, Kim SK, Vogel H, Wood LD, Hruban RH, Curtis C, Attardi LD. A p53 Super-tumor Suppressor Reveals a Tumor Suppressive p53-Ptpn14-Yap Axis in Pancreatic Cancer. Cancer Cell 2017; 32:460-473.e6. [PMID: 29017057 PMCID: PMC5659188 DOI: 10.1016/j.ccell.2017.09.007] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/19/2017] [Accepted: 09/08/2017] [Indexed: 12/25/2022]
Abstract
The p53 transcription factor is a critical barrier to pancreatic cancer progression. To unravel mechanisms of p53-mediated tumor suppression, which have remained elusive, we analyzed pancreatic cancer development in mice expressing p53 transcriptional activation domain (TAD) mutants. Surprisingly, the p5353,54 TAD2 mutant behaves as a "super-tumor suppressor," with an enhanced capacity to both suppress pancreatic cancer and transactivate select p53 target genes, including Ptpn14. Ptpn14 encodes a negative regulator of the Yap oncoprotein and is necessary and sufficient for pancreatic cancer suppression, like p53. We show that p53 deficiency promotes Yap signaling and that PTPN14 and TP53 mutations are mutually exclusive in human cancers. These studies uncover a p53-Ptpn14-Yap pathway that is integral to p53-mediated tumor suppression.
Collapse
Affiliation(s)
- Stephano S Mello
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liz J Valente
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nitin Raj
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jose A Seoane
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brittany M Flowers
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jacob McClendon
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kathryn T Bieging-Rolett
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jonghyeob Lee
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Danton Ivanochko
- Princess Margaret Cancer Centre, Structural Genomics Consortium and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Margaret M Kozak
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daniel T Chang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Teri A Longacre
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Albert C Koong
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Cheryl H Arrowsmith
- Princess Margaret Cancer Centre, Structural Genomics Consortium and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Seung K Kim
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hannes Vogel
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laura D Wood
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christina Curtis
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laura D Attardi
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Chen MJ, Dixon JE, Manning G. Genomics and evolution of protein phosphatases. Sci Signal 2017; 10:10/474/eaag1796. [DOI: 10.1126/scisignal.aag1796] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
23
|
Wilson KE, Yang N, Mussell AL, Zhang J. The Regulatory Role of KIBRA and PTPN14 in Hippo Signaling and Beyond. Genes (Basel) 2016; 7:genes7060023. [PMID: 27240404 PMCID: PMC4929422 DOI: 10.3390/genes7060023] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/17/2016] [Accepted: 05/19/2016] [Indexed: 12/21/2022] Open
Abstract
The Hippo signaling pathway regulates cellular proliferation and survival, thus exerting profound effects on normal cell fate and tumorigenesis. Pivotal effectors of this pathway are YAP/TAZ, transcriptional co-activators whose dysfunction contributes to the development of cancer. Complex networks of intracellular and extracellular signaling pathways that modulate YAP and TAZ activities have recently been identified. Among them, KIBRA and PTPN14 are two evolutionarily-conserved and important YAP/TAZ upstream regulators. They can negatively regulate YAP/TAZ functions separately or in concert. In this review, we summarize the current and emerging regulatory roles of KIBRA and PTPN14 in the Hippo pathway and their functions in cancer.
Collapse
Affiliation(s)
- Kayla E Wilson
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| | - Nuo Yang
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| | - Ashley L Mussell
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| | - Jianmin Zhang
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| |
Collapse
|
24
|
Miller CL, Muthupalani S, Shen Z, Drees F, Ge Z, Feng Y, Chen X, Gong G, Nagar KK, Wang TC, Gertler FB, Fox JG. Lamellipodin-Deficient Mice: A Model of Rectal Carcinoma. PLoS One 2016; 11:e0152940. [PMID: 27045955 PMCID: PMC4821566 DOI: 10.1371/journal.pone.0152940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/20/2016] [Indexed: 01/27/2023] Open
Abstract
During a survey of clinical rectal prolapse (RP) cases in the mouse population at MIT animal research facilities, a high incidence of RP in the lamellipodin knock-out strain, C57BL/6-Raph1tm1Fbg (Lpd-/-) was documented. Upon further investigation, the Lpd-/- colony was found to be infected with multiple endemic enterohepatic Helicobacter species (EHS). Lpd-/- mice, a transgenic mouse strain produced at MIT, have not previously shown a distinct immune phenotype and are not highly susceptible to other opportunistic infections. Predominantly male Lpd-/- mice with RP exhibited lesions consistent with invasive rectal carcinoma concomitant to clinically evident RP. Multiple inflammatory cytokines, CD11b+Gr1+ myeloid-derived suppressor cell (MDSC) populations, and epithelial cells positive for a DNA damage biomarker, H2AX, were elevated in affected tissue, supporting their role in the neoplastic process. An evaluation of Lpd-/- mice with RP compared to EHS-infected, but clinically normal (CN) Lpd-/- animals indicated that all of these mice exhibit some degree of lower bowel inflammation; however, mice with prolapses had significantly higher degree of focal lesions at the colo-rectal junction. When Helicobacter spp. infections were eliminated in Lpd-/- mice by embryo transfer rederivation, the disease phenotype was abrogated, implicating EHS as a contributing factor in the development of rectal carcinoma. Here we describe lesions in Lpd-/- male mice consistent with a focal inflammation-induced neoplastic transformation and propose this strain as a mouse model of rectal carcinoma.
Collapse
Affiliation(s)
- Cassandra L. Miller
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Sureshkumar Muthupalani
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Zeli Shen
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Frauke Drees
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Zhongming Ge
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Yan Feng
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Xiaowei Chen
- Division of Digestive and Liver Diseases, Columbia University, New York, NY, United States of America
| | - Guanyu Gong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Karan K. Nagar
- Division of Digestive and Liver Diseases, Columbia University, New York, NY, United States of America
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Columbia University, New York, NY, United States of America
| | - Frank B. Gertler
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
25
|
Chen KE, Li MY, Chou CC, Ho MR, Chen GC, Meng TC, Wang AJ. Substrate Specificity and Plasticity of FERM-Containing Protein Tyrosine Phosphatases. Structure 2015; 23:653-64. [DOI: 10.1016/j.str.2015.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 01/18/2015] [Accepted: 01/24/2015] [Indexed: 10/23/2022]
|
26
|
Belle L, Ali N, Lonic A, Li X, Paltridge JL, Roslan S, Herrmann D, Conway JRW, Gehling FK, Bert AG, Crocker LA, Tsykin A, Farshid G, Goodall GJ, Timpson P, Daly RJ, Khew-Goodall Y. The tyrosine phosphatase PTPN14 (Pez) inhibits metastasis by altering protein trafficking. Sci Signal 2015; 8:ra18. [PMID: 25690013 DOI: 10.1126/scisignal.2005547] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Factors secreted by tumor cells shape the local microenvironment to promote invasion and metastasis, as well as condition the premetastatic niche to enable secondary-site colonization and growth. In addition to this secretome, tumor cells have increased abundance of growth-promoting receptors at the cell surface. We found that the tyrosine phosphatase PTPN14 (also called Pez, which is mutated in various cancers) suppressed metastasis by reducing intracellular protein trafficking through the secretory pathway. Knocking down PTPN14 in tumor cells or injecting the peritoneum of mice with conditioned medium from PTPN14-deficient cell cultures promoted the growth and metastasis of breast cancer xenografts. Loss of catalytically functional PTPN14 increased the secretion of growth factors and cytokines, such as IL-8 (interleukin-8), and increased the abundance of EGFR (epidermal growth factor receptor) at the cell surface of breast cancer cells and of FLT4 (vascular endothelial growth factor receptor 3) at the cell surface of primary lymphatic endothelial cells. We identified RIN1 (Ras and Rab interactor 1) and PRKCD (protein kinase C-δ) as binding partners and substrates of PTPN14. Similar to cells overexpressing PTPN14, receptor trafficking to the cell surface was inhibited in cells that lacked PRKCD or RIN1 or expressed a nonphosphorylatable RIN1 mutant, and cytokine secretion was decreased in cells treated with PRKCD inhibitors. Invasive breast cancer tissue had decreased expression of PTPN14, and patient survival was worse when tumors had increased expression of the genes encoding RIN1 or PRKCD. Thus, PTPN14 prevents metastasis by restricting the trafficking of both soluble and membrane-bound proteins.
Collapse
Affiliation(s)
- Leila Belle
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia. Discipline of Biochemistry, School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Naveid Ali
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Ana Lonic
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia. Department of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Xiaochun Li
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia
| | - James L Paltridge
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia. Discipline of Biochemistry, School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Suraya Roslan
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia
| | - David Herrmann
- Cancer Research Program, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - James R W Conway
- Cancer Research Program, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Freya K Gehling
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia
| | - Andrew G Bert
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia
| | - Lesley A Crocker
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia
| | - Anna Tsykin
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia
| | - Gelareh Farshid
- Division of Tissue Pathology, SA Pathology, Adelaide, South Australia 5000, Australia
| | - Gregory J Goodall
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia. Department of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia. School and Molecular and Biomedical Science, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Paul Timpson
- Cancer Research Program, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
| | - Roger J Daly
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia. Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Yeesim Khew-Goodall
- Centre for Cancer Biology, an Alliance between SA Pathology and University of South Australia, Adelaide, South Australia 5000, Australia. Discipline of Biochemistry, School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, South Australia 5005, Australia. Department of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia.
| |
Collapse
|
27
|
Yuan L, Liu X. Platelets are associated with xenograft tumor growth and the clinical malignancy of ovarian cancer through an angiogenesis-dependent mechanism. Mol Med Rep 2014; 11:2449-58. [PMID: 25502723 PMCID: PMC4337475 DOI: 10.3892/mmr.2014.3082] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 11/05/2014] [Indexed: 12/13/2022] Open
Abstract
Platelets are known to facilitate tumor metastasis and thrombocytosis has been associated with an adverse prognosis in ovarian cancer. However, the role of platelets in primary tumour growth remains to be elucidated. The present study demonstrated that the expression levels of various markers in platelets, endothelial adherence and angiogenesis, including, platelet glycoprotein IIb (CD41), platelet endothelial cell adhesion molecule 1 (CD31), vascular endothelial growth factor (VEGF), lysyl oxidase, focal adhesion kinase and breast cancer anti-estrogen resistance 1, were expressed at higher levels in patients with malignant carcinoma, compared with those with borderline cystadenoma and cystadenoma. In addition, the endothelial markers CD31 and VEGF were found to colocalize with the platelet marker CD41 in the malignant samples. Since mice transplanted with human ovarian cancer cells (SKOV3) demonstrated elevated tumor size and decreased survival rate when treated with thrombin or thrombopoietin (TPO), the platelets appeared to promote primary tumor growth. Depleting platelets using antibodies or by pretreating the cancer cells with hirudin significantly attenuated the transplanted tumor growth. The platelets contributed to late, but not early stages of tumor proliferation, as mice treated with platelet-depleting antibody 1 day prior to and 11 days after tumor transplantation had the same tumor volumes. By contrast, tumor size in the early TPO-injected group was increased significantly compared with the late TPO-injected group. These findings suggested that the interplay between platelets and angiogenesis may contribute to ovarian cancer growth. Therefore, platelets and their associated signaling and adhesive molecules may represent potential therapeutic targets for ovarian cancer.
Collapse
Affiliation(s)
- Lei Yuan
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| | - Xishi Liu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
| |
Collapse
|
28
|
Seo M, Lee S, Kim JH, Lee WH, Hu G, Elledge SJ, Suk K. RNAi-based functional selection identifies novel cell migration determinants dependent on PI3K and AKT pathways. Nat Commun 2014; 5:5217. [PMID: 25347953 PMCID: PMC6581447 DOI: 10.1038/ncomms6217] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 09/09/2014] [Indexed: 12/12/2022] Open
Abstract
Lentiviral short hairpin RNA (shRNA)-mediated genetic screening is a powerful tool for identifying loss-of-function phenotype in mammalian cells. Here, we report the identification of 91 cell migration-regulating genes using unbiased genome-wide functional genetic selection. Individual knockdown or cDNA overexpression of a set of 10 candidates reveals that most of these cell migration determinants are strongly dependent on the PI3K/PTEN/AKT pathway and on their downstream signals, such as FOXO1 and p70S6K1. ALK, one of the cell migration promoting genes, uniquely uses p55γ regulatory subunit of PI3K, rather than more common p85 subunit, to trigger the activation of the PI3K-AKT pathway. Our method enables the rapid and cost-effective genome-wide selection of cell migration regulators. Our results emphasize the importance of the PI3K/PTEN/AKT pathway as a point of convergence for multiple regulators of cell migration.
Collapse
Affiliation(s)
- Minchul Seo
- 1] Department of Pharmacology, Brain Science &Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea [2] College of Medicine, Dongguk University, Gyeongju, Republic of Korea
| | - Shinrye Lee
- 1] Department of Pharmacology, Brain Science &Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea [2] Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Jong-Heon Kim
- Department of Pharmacology, Brain Science &Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Won-Ha Lee
- KNU Creative BioResearch Group, School of Life Sciences and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| | - Guang Hu
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health and Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Stephen J Elledge
- Department of Genetics, Howard Hughes Medical Institute, Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science &Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
29
|
Zhao S, Sedwick D, Wang Z. Genetic alterations of protein tyrosine phosphatases in human cancers. Oncogene 2014; 34:3885-94. [PMID: 25263441 PMCID: PMC4377308 DOI: 10.1038/onc.2014.326] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 08/20/2014] [Accepted: 08/21/2014] [Indexed: 12/12/2022]
Abstract
Protein tyrosine phosphatases (PTPs) are enzymes that remove phosphate from tyrosine residues in proteins. Recent whole-exome sequencing of human cancer genomes reveals that many PTPs are frequently mutated in a variety of cancers. Among these mutated PTPs, protein tyrosine phosphatase T (PTPRT) appears to be the most frequently mutated PTP in human cancers. Beside PTPN11 which functions as an oncogene in leukemia, genetic and functional studies indicate that most of mutant PTPs are tumor suppressor genes. Identification of the substrates and corresponding kinases of the mutant PTPs may provide novel therapeutic targets for cancers harboring these mutant PTPs.
Collapse
Affiliation(s)
- S Zhao
- 1] Division of Gastroenterology and Hepatology and Shanghai Institution of Digestive Disease, Shanghai Jiao-Tong University School of Medicine Renji Hospital, Shanghai, China [2] Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA [3] Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - D Sedwick
- 1] Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA [2] Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Z Wang
- 1] Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA [2] Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
30
|
Lee H, Yi JS, Lawan A, Min K, Bennett AM. Mining the function of protein tyrosine phosphatases in health and disease. Semin Cell Dev Biol 2014; 37:66-72. [PMID: 25263013 DOI: 10.1016/j.semcdb.2014.09.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 09/21/2014] [Indexed: 12/31/2022]
Abstract
Protein tyrosine phosphatases (PTPs) play a crucial role in the regulation of human health and it is now clear that PTP dysfunction is causal to a variety of human diseases. Research in the PTP field has accelerated dramatically over the last decade fueled by cutting-edge technologies in genomic and proteomic techniques. This system-wide non-biased approach when applied to the discovery of PTP function has led to the elucidation of new and unanticipated roles for the PTPs. These discoveries, driven by genomic and proteomic approaches, have uncovered novel PTP findings that range from those that describe fundamental cell signaling mechanisms to implications for PTPs as novel therapeutic targets for the treatment of human disease. This review will discuss how new PTP functions have been uncovered through studies that have utilized genomic and proteomic technologies and strategies.
Collapse
Affiliation(s)
- Hojin Lee
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Jae-Sung Yi
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Ahmed Lawan
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Kisuk Min
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Anton M Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
31
|
Nikonova AS, Gaponova AV, Kudinov AE, Golemis EA. CAS proteins in health and disease: an update. IUBMB Life 2014; 66:387-95. [PMID: 24962474 DOI: 10.1002/iub.1282] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 06/07/2014] [Indexed: 12/30/2022]
Abstract
The CAS family of scaffolding proteins has increasingly attracted scrutiny as important for regulation of cancer-associated signaling. BCAR1 (also known as p130Cas), NEDD9 (HEF1, Cas-L), EFS (Sin), and CASS4 (HEPL) are regulated by and mediate cell attachment, growth factor, and chemokine signaling. Altered expression and activity of CAS proteins are now known to promote metastasis and drug resistance in cancer, influence normal development, and contribute to the pathogenesis of heart and pulmonary disease. In this article, we provide an update on recently published studies describing signals regulating and regulated by CAS proteins, and evidence for biological activity of CAS proteins in normal development, cancer, and other pathological conditions.
Collapse
Affiliation(s)
- Anna S Nikonova
- Program in Developmental Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
32
|
Wang X, Ji X, Chen J, Yan D, Zhang Z, Wang Q, Xi X, Feng Y. SOX2 enhances the migration and invasion of ovarian cancer cells via Src kinase. PLoS One 2014; 9:e99594. [PMID: 24937695 PMCID: PMC4061006 DOI: 10.1371/journal.pone.0099594] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 05/15/2014] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is the leading cause of death among gynecologic cancers and is the fifth leading cause of all cancer-related deaths among women. The development of novel molecular targets is therefore important to many patients. Recently, the SRY-related transcription factor SOX2 has been widely reported to be involved in multiple pathophysiological diseases, including maintenance of stem cell characteristics and carcinogenesis. Up to now, SOX2 has been mainly shown to promote the development of cancer, although its inhibitory roles in cancer have also been reported. However, the role of SOX2 in ovarian cancer is largely unknown. In the present study, we detected the expression of SOX2 in 64 human serous ovarian carcinoma (SOC) tissues and paired corresponding metastatic specimens using immunohistochemistry. The results showed that the expression of SOX2 in primary tumors is much lower than that in the corresponding metastatic lesions. We further found that SOX2 overexpression promotes proliferation, migration and invasion, while inhibiting adhesion abilities of SOC cells. Finally, we found that SOX2 targets Src kinase, a non-receptor tyrosine kinase that regulates cell migration, invasion and adhesion in SOC cells. Together, these results suggested that Src kinase is a key molecule in SOX2-mediated migration and invasion of SOC cells.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Xiaoning Ji
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Jiazhou Chen
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Dong Yan
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Zhenbo Zhang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Qifeng Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaowei Xi
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Youji Feng
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| |
Collapse
|
33
|
Selner NG, Luechapanichkul R, Chen X, Neel BG, Zhang ZY, Knapp S, Bell CE, Pei D. Diverse levels of sequence selectivity and catalytic efficiency of protein-tyrosine phosphatases. Biochemistry 2014; 53:397-412. [PMID: 24359314 PMCID: PMC3954597 DOI: 10.1021/bi401223r] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The sequence selectivity of 14 classical protein-tyrosine phosphatases (PTPs) (PTPRA, PTPRB, PTPRC, PTPRD, PTPRO, PTP1B, SHP-1, SHP-2, HePTP, PTP-PEST, TCPTP, PTPH1, PTPD1, and PTPD2) was systematically profiled by screening their catalytic domains against combinatorial peptide libraries. All of the PTPs exhibit similar preference for pY peptides rich in acidic amino acids and disfavor positively charged sequences but differ vastly in their degrees of preference/disfavor. Some PTPs (PTP-PEST, SHP-1, and SHP-2) are highly selective for acidic over basic (or neutral) peptides (by >10(5)-fold), whereas others (PTPRA and PTPRD) show no to little sequence selectivity. PTPs also have diverse intrinsic catalytic efficiencies (kcat/KM values against optimal substrates), which differ by >10(5)-fold due to different kcat and/or KM values. Moreover, PTPs show little positional preference for the acidic residues relative to the pY residue. Mutation of Arg47 of PTP1B, which is located near the pY-1 and pY-2 residues of a bound substrate, decreased the enzymatic activity by 3-18-fold toward all pY substrates containing acidic residues anywhere within the pY-6 to pY+5 region. Similarly, mutation of Arg24, which is situated near the C-terminus of a bound substrate, adversely affected the kinetic activity of all acidic substrates. A cocrystal structure of PTP1B bound with a nephrin pY(1193) peptide suggests that Arg24 engages in electrostatic interactions with acidic residues at the pY+1, pY+2, and likely other positions. These results suggest that long-range electrostatic interactions between positively charged residues near the PTP active site and acidic residues on pY substrates allow a PTP to bind acidic substrates with similar affinities, and the varying levels of preference for acidic sequences by different PTPs are likely caused by the different electrostatic potentials near their active sites. The implications of the varying sequence selectivity and intrinsic catalytic activities with respect to PTP in vivo substrate specificity and biological functions are discussed.
Collapse
Affiliation(s)
- Nicholas G. Selner
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, OH 43210, USA
| | - Rinrada Luechapanichkul
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, OH 43210, USA
| | - Xianwen Chen
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, OH 43210, USA
| | - Benjamin G. Neel
- Princess Margaret Cancer Center, University Health Network, and Department of Medical Biophysics, University of Toronto, 610 University Avenue, Room 7-504, Toronto, ON M5G 2M9, Canada
| | - Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stefan Knapp
- Structural Genomics Consortium and Target Discovery Institute, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Charles E. Bell
- Department of Molecular and Cellular Biochemistry, The Ohio State University, 1645 Neil Avenue, Columbus, OH 43210
| | - Dehua Pei
- Department of Chemistry and Biochemistry, The Ohio State University, 484 West 12 Avenue, Columbus, OH 43210, USA
| |
Collapse
|
34
|
Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe 2013; 14:195-206. [PMID: 23954158 PMCID: PMC3770529 DOI: 10.1016/j.chom.2013.07.012] [Citation(s) in RCA: 1464] [Impact Index Per Article: 133.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 06/14/2013] [Accepted: 07/17/2013] [Indexed: 02/08/2023]
Abstract
Fusobacterium nucleatum (Fn) has been associated with colorectal cancer (CRC), but causality and underlying mechanisms remain to be established. We demonstrate that Fn adheres to, invades, and induces oncogenic and inflammatory responses to stimulate growth of CRC cells through its unique FadA adhesin. FadA binds to E-cadherin, activates β-catenin signaling, and differentially regulates the inflammatory and oncogenic responses. The FadA-binding site on E-cadherin is mapped to an 11-amino-acid region. A synthetic peptide derived from this region of E-cadherin abolishes FadA-induced CRC cell growth and oncogenic and inflammatory responses. The fadA gene levels in the colon tissue from patients with adenomas and adenocarcinomas are >10-100 times higher compared to normal individuals. The increased FadA expression in CRC correlates with increased expression of oncogenic and inflammatory genes. This study unveils a mechanism by which Fn can drive CRC and identifies FadA as a potential diagnostic and therapeutic target for CRC.
Collapse
Affiliation(s)
| | - Xiaowei Wang
- Department of Periodontics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Wendy Liu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Yujun Hao
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Guifang Cai
- School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Yiping W. Han
- Department of Periodontics, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| |
Collapse
|