1
|
Jiang C, Liu R, Chang Y, Zhang S, Li X, Zhao Z, Quan M, Wang Q, Zhou H, Hou X, Fang H. Design and synthesis of novel benzoic acid derivatives as striatal-enriched protein tyrosine phosphatase (STEP) inhibitors with neuroprotective properties. Eur J Med Chem 2025; 283:117135. [PMID: 39657460 DOI: 10.1016/j.ejmech.2024.117135] [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: 08/12/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
As a central nervous system-specific member of the protein tyrosine phosphatase (PTP) family, the striatal-enriched protein tyrosine phosphatase (STEP) is an attractive drug target for neurodegenerative diseases. Here, we reported the discovery of a series of benzoic acid derivatives as new STEP inhibitors. Among them, compound 14b exhibited good STEP inhibitory activity and displayed selectivity against other PTPs. The neuroprotective activity of compound 14b was evaluated against glutamate-induced oxidative cell death in HT22 cells. Results indicated that compound 14b co-treatment prevented cell death and reduced cellular ROS accumulation. Compound 14b inhibited cell apoptosis by upregulating BCL-2 expression and downregulating BAX and C-caspase3 expression. Moreover, compound 14b was also found to provide neuroprotection to primary cortical neurons after oxygen-glucose deprivation/reoxygenation (OGD/R). Further structural elaboration of compound 14b may provide new drug candidates for neurodegenerative diseases.
Collapse
Affiliation(s)
- Chunxue Jiang
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Renshuai Liu
- Orthopaedic Research Center of Shandong University, Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yong Chang
- Orthopaedic Research Center of Shandong University, Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Shiji Zhang
- Orthopaedic Research Center of Shandong University, Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xue Li
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Zhongcheng Zhao
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Mengyao Quan
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Quande Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Normal University, Guilin, 541004, China
| | - Hengxing Zhou
- Orthopaedic Research Center of Shandong University, Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Xuben Hou
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Hao Fang
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Normal University, Guilin, 541004, China.
| |
Collapse
|
2
|
Johnson D, Dixit M, Kirubakaran S. Biochemical and Structural Studies of Protein Tyrosine Phosphatase PTP-PEST (PTPN12) in Search of Small Molecule Inhibitors. Chem Biol Drug Des 2025; 105:e70058. [PMID: 39895370 DOI: 10.1111/cbdd.70058] [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: 06/25/2024] [Revised: 12/17/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
PTP-PEST (also known as PTPN12) regulates cellular signaling and transduction pathways by dephosphorylating its substrate. PTP-PEST is considered an important drug target owing to its involvement in cancer progression and myocardial injury. Till now only a few inhibitors are currently being studied in the inhibition of PTP-PEST, majorly belonging to the class of metal-based drugs. In this study, we aimed to investigate small molecules that could potentially inhibit PTP-PEST for further development of PTP-PEST inhibitors. As an approach, we used an in silico molecular docking technique to screen an in-house synthesized molecular library. Further, we validated the docking results by in vitro inhibition screening of the best molecules using the purified catalytic domain of human PTP-PEST, which was over-expressed in E.coli. We identified a myo-inositol based derivative, J1-65, which binds to PTP-PEST and results in the competitive inhibition of the protein. Further, we confirmed this protein-ligand binding using binding affinity studies based on protein thermal shift assay and in silico molecular dynamic simulations. Our efforts to discover a novel scaffold for inhibiting hPTP-PEST mark a crucial stride in laying the groundwork for the future development of selective PTP-PEST inhibitors.
Collapse
Affiliation(s)
- Delna Johnson
- Department of Chemistry, Indian Institute of Technology, Gandhinagar, Gujarat, India
| | - Madhulika Dixit
- Centre of Excellence (CoE) in Molecular Medicine, Department of Biotechnology, Indian Institute of Technology, Chennai, Tamil Nadu, India
| | - Sivapriya Kirubakaran
- Department of Chemistry, Indian Institute of Technology, Gandhinagar, Gujarat, India
| |
Collapse
|
3
|
Thirumalai Srinivasan S, Manikandan A, Manoj N, Dixit M, Vemparala S. Role of Tyrosine Phosphorylation in PTP-PEST. J Phys Chem B 2024; 128:10581-10592. [PMID: 39423851 DOI: 10.1021/acs.jpcb.4c04047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
We study the influence of tyrosine phosphorylation on PTP-PEST, a cytosolic protein tyrosine phosphatase. Utilizing a combination of experimental data and computational modeling, specific tyrosine sites, notably, Y64 and Y88, are identified for potential phosphorylation. Phosphorylation at these sites affects loop dynamics near the catalytic site, altering interactions among key residues and modifying the size of the binding pocket. This, in turn, impacts substrate binding, as indicated by changes in the binding energy. Our findings provide insights into the structural and functional consequences of tyrosine phosphorylation on PTP-PEST, enhancing our understanding of its effects on substrate binding and catalytic conformation.
Collapse
Affiliation(s)
| | - Amrutha Manikandan
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, India
| | - Narayanan Manoj
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, India
| | - Madhulika Dixit
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, India
| | - Satyavani Vemparala
- Homi Bhabha National Institute, Mumbai 400094, India
- The Institute of Mathematical Sciences, Chennai 600113, India
| |
Collapse
|
4
|
Cheng X. A Comprehensive Review of HER2 in Cancer Biology and Therapeutics. Genes (Basel) 2024; 15:903. [PMID: 39062682 PMCID: PMC11275319 DOI: 10.3390/genes15070903] [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: 06/01/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2), a targetable transmembrane glycoprotein receptor of the epidermal growth factor receptor (EGFR) family, plays a crucial role in cell proliferation, survival, and differentiation. Aberrant HER2 signaling is implicated in various cancers, particularly in breast and gastric cancers, where HER2 overexpression or amplification correlates with aggressive tumor behavior and poor prognosis. HER2-activating mutations contribute to accelerated tumorigenesis and metastasis. This review provides an overview of HER2 biology, signaling pathways, mechanisms of dysregulation, and diagnostic approaches, as well as therapeutic strategies targeting HER2 in cancer. Understanding the intricate details of HER2 regulation is essential for developing effective targeted therapies and improving patient outcomes.
Collapse
Affiliation(s)
- Xiaoqing Cheng
- Department of Oncology, School of Medicine, Washington University in Saint Louis, Saint Louis, MO 63108, USA
| |
Collapse
|
5
|
O’Neill CE, Sun K, Sundararaman S, Chang JC, Glynn SA. The impact of nitric oxide on HER family post-translational modification and downstream signaling in cancer. Front Physiol 2024; 15:1358850. [PMID: 38601214 PMCID: PMC11004480 DOI: 10.3389/fphys.2024.1358850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/16/2024] [Indexed: 04/12/2024] Open
Abstract
The human epidermal growth factor receptor (HER) family consists of four members, activated by two families of ligands. They are known for mediating cell-cell interactions in organogenesis, and their deregulation has been associated with various cancers, including breast and esophageal cancers. In particular, aberrant epidermal growth factor receptor (EGFR) and HER2 signaling drive disease progression and result in poorer patient outcomes. Nitric oxide (NO) has been proposed as an alternative activator of the HER family and may play a role in this aberrant activation due to its ability to induce s-nitrosation and phosphorylation of the EGFR. This review discusses the potential impact of NO on HER family activation and downstream signaling, along with its role in the efficacy of therapeutics targeting the family.
Collapse
Affiliation(s)
- Ciara E. O’Neill
- Lambe Institute for Translational Research, Discipline of Pathology, School of Medicine, University of Galway, Galway, Ireland
| | - Kai Sun
- Houston Methodist Research Institute, Houston, TX, United States
- Dr Mary and Ron Neal Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| | | | - Jenny C. Chang
- Houston Methodist Research Institute, Houston, TX, United States
- Dr Mary and Ron Neal Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| | - Sharon A. Glynn
- Lambe Institute for Translational Research, Discipline of Pathology, School of Medicine, University of Galway, Galway, Ireland
| |
Collapse
|
6
|
Lan L, Cao H, Zhao L, Cui W, Wang B. PTPN12 down-regulated by miR-146b-3p gene affects the malignant progression of laryngeal squamous cell carcinoma. Open Med (Wars) 2023; 18:20230727. [PMID: 37333450 PMCID: PMC10276617 DOI: 10.1515/med-2023-0727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 03/31/2023] [Accepted: 05/07/2023] [Indexed: 06/20/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a common malignancy among men in the anatomical position of head and neck. Hoarseness, pharyngalgia, and dyspnea are common symptoms. LSCC is a complex polygenic carcinoma that is caused by many factors involving polygenic alteration, environmental pollution, tobacco, and human papillomavirus. Classical protein tyrosine phosphatase nonreceptor type 12 (PTPN12) has been extensively studied to decipher its mechanism as a tumor suppressor gene in various human carcinomas; however, there is no comprehensive elucidation of the PTPN12 expression and its regulatory mechanisms in LSCC. As such, we expect to provide new insights for finding new biomarkers and effective therapeutic targets in LSCC. Immunohistochemical staining, western blot (WB), and quantitative real-time RT-PCR (qRT-PCR) were used for the messenger RNA (mRNA) and protein expression analyses of PTPN12, respectively. 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, clone formation, transwell migration, and transwell invasion assays were used to assess the proliferation, migration, and invasion ability of LSCC cells. Online prediction and design software tools (http://www.targetscan.org/ and http://www.microRNA.org) were used to predict associated miRNA. Studying the targeted regulatory relationship between miR-146b-3p and PTPN12 was based on dual luciferase reporter gene analysis. qRT-PCR was used to assess miR-146b-3p expression in LSCC. miR-146b-3p inhibitor and mimic were transfected, followed by qRT-PCR and WB assays to detect the expression of PTPN12. The gain and loss functional experiments were used to investigate the effects of miR-146b-3p transfection on the proliferation, migration, and invasion of tumor cells. Online bioinformatics prediction software (https://cn.string-db.org/ and https://www.genecards.org/) was used to determine potential downstream target genes of PTPN12. qRT-PCR and WB analyses were used to assess the mRNA and protein expression levels of target genes. Our study showed significantly decreased mRNA and protein expression levels of PTPN12 in LSCC compared with the adjacent normal tissues. The lower PTPN12 mRNA expression was correlated with pathological differentiation, and lower PTPN12 protein expression was correlated with the TNM stage in LSCC tissues. The subsequent in vitro functional analyses showed the inhibitory effect of PTPN12 over-expression on the proliferation, migration, and invasiveness abilities of LSCC cell line. Using online prediction and design software, miR-146b-3p was searched to target PTPN12. The miR-146b-3p was expressed at a high level in LSCC tissues and cell lines. Luciferase reporter assay exhibited that miR-146b-3p inhibited the luciferase activity of PTPN12 markedly. The functional analyses showed the tumor-promoting role of miR-146b-3p on the proliferation, migration, and invasiveness abilities of LSCC cell. Furthermore, co-transfection of cells with miR-146b-3p and PTPN12 significantly restored the inhibitory effect of PTPN12 on LSCC cell growth, migration, and invasiveness. This phenomenon unveiled that miR-146b-3p regulated the proliferation, migration, and invasion of LSCC cells by targeting PTPN12. EGFR and ERBB2 were selected as the downstream-regulation target genes. Up-regulation of PTPN12 significantly suppressed EGFR expression. Accordingly, the miR-146b-3p mimic significantly up-regulated the EGFR expression. However, up-regulation of PTPN12 and miR-146b-3p mimic suppressed ERBB2 protein expression but induced its gene expression. Down-regulation of PTPN12 is associated with up-regulation of miR-146b-3p in LSCC. Moreover, PTPN12 serves as a tumor suppressor gene through regulating the proliferation, migration, and invasion of LSCC cells. miR-146b-3p/PTPN12 axis is expected to be a novel therapeutic target in LSCC.
Collapse
Affiliation(s)
- Lili Lan
- Otolaryngology Head and Neck Surgery Department, The Second Hospital of Hebei Medical University, Shijiazhuang050005, Hebei, China
- Otolaryngology Head and Neck Surgery Department, The Fourth Hospital of Hebei Medical University, Shijiazhuang050011, Hebei, China
| | - Huan Cao
- Otolaryngology Head and Neck Surgery Department, The Second Hospital of Hebei Medical University, Shijiazhuang050005, Hebei, China
| | - Lei Zhao
- Otolaryngology Head and Neck Surgery Department, The Second Hospital of Hebei Medical University, Shijiazhuang050005, Hebei, China
| | - Weina Cui
- Otolaryngology Head and Neck Surgery Department, The Second Hospital of Hebei Medical University, Shijiazhuang050005, Hebei, China
| | - Baoshan Wang
- Otolaryngology Head and Neck Surgery Department, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang050005, Hebei, China
| |
Collapse
|
7
|
Wang X, Wang X, Lai J, Xu W, Zhu W, Chen G. Protein tyrosine phosphatase non-receptor type 12 suppresses tumor progression in osteosarcoma cells. J Orthop Sci 2023; 28:468-475. [PMID: 35063332 DOI: 10.1016/j.jos.2021.12.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/15/2021] [Accepted: 12/22/2021] [Indexed: 02/09/2023]
Abstract
BACKGROUND Protein tyrosine phosphatase non-receptor 12 (PTPN12) plays a prominent role in various cancers as a tumor suppressor. However, the expression of PTPN12 and its biological functions in osteosarcoma (OS) remains to be determined. METHODS PTPN12 expression in OS was explored in public databases and detected by immunohistochemistry and Western blot. The cell viability was determined by Cell Counting Kit-8 (CCK-8) assay and colony formation. The cell migration and invasion were assessed by the Transwell assay. Flow cytometry analysis was applied to detect cell apoptosis and cell cycle distribution. To investigate the related mechanism, the levels of EGFR and downstream proteins were detected by Western blot. RESULTS PTPN12 expression was significantly decreased in OS samples in GEO database and our hospital. OS cell lines in Cancer Cell Line Encyclopedia (CCLE) database and our cultured OS cells also demonstrated low PTPN12 expression. Lentivirus-induced overexpression of PTPN12 significantly inhibited the cell viability, migration and invasion of 143B and U2OS cells. The results of flow cytometry found that PTPN12 overexpression promoted cell apoptosis and induced cell cycle arrest at G1 phase in 143B and U2OS cells. The phosphorylation levels of EGFR and subsequent proteins of the PI3K/AKT and ERK pathways were inactivated as a result of PTPN12 overexpression in OS. CONCLUSION PTPN12 plays a tumor suppressive role in OS cells. Restoring of PTPN12 activity may provide new insights for the treatment of this disease.
Collapse
Affiliation(s)
- Xinwu Wang
- Department of Orthopaedics, The First Hospital of Putian City, Putian, Fujian, 351199, China
| | - Xinwen Wang
- Department of Orthopaedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, 350005, China
| | - Jiankun Lai
- Department of Orthopaedics, Dongguan People 's Hospital, Dongguan, Guangdong, 523059, China
| | - Weifeng Xu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Wenxiong Zhu
- Department of Orthopaedics, Dongguan People 's Hospital, Dongguan, Guangdong, 523059, China.
| | - Guoxian Chen
- Department of Orthopaedics, The First Hospital of Putian City, Putian, Fujian, 351199, China.
| |
Collapse
|
8
|
Hu D, Tian Y, Ye L, Xin Y, Shao J. Transthyretin-induced increase in circ_0007411 represses neovascularization of human retinal microvascular endothelial cells in hyperglycemia via the miR-548m/PTPN12/SKP1/EGFR pathway. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:556. [PMID: 35722376 PMCID: PMC9201143 DOI: 10.21037/atm-22-1276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/27/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND To investigate the mechanism of transthyretin (TTR) induced high expression of circ_0007411 and its parent gene, protein tyrosine phosphatase nonreceptor type 12 (PTPN12) in human retinal microvascular endothelial cells (hRECs) cultivated under high glucose condition. METHODS The levels of PTPN12, circ_0007411, miR-548m, S-phase kinase associated protein 1 (SKP1) and epidermal growth factor receptor (EGFR) were detected by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The direct interaction between circ_0007411/PTPN12 and miR-548m was investigated via Dual-luciferase reporter assay. The physiological characterization of hRECs was investigated through Cell Counting Kit-8 (CCK-8), 5-Ethynyl-2'-deoxyuridine (EdU) labelling, Transwell, flow cytometry (FCM), wound healing, and tube formation assays. Co-immunoprecipitation (Co-IP) was used to detect the interaction between PTPN12 and SKP1. The function of PTPN12 against diabetic retinopathy (DR) was studied in streptozotocin (STZ) induced DR C57BL/6 mice. RESULTS The levels of circ_0007411 was increased in hRECs in hyperglycemia with the induction of TTR. The overexpressed circ_0007411 could significantly enhance the level of PTPN12 and repress that of miR-548m, and it could enhance apoptosis and prohibit the proliferation, migration, and tube formation of hRECs. miR-548m mimics enhanced the proliferation, migration, and tube formation of hRECs by reducing the expression level of PTPN12 and promoting that of EGFR, whereas circ_0007411 rescued it. The direct binding of PTPN12 and SKP1 was confirmed by Co-IP. Additionally, the anti-neovascularization function of PTPN12 was confirmed in a STZ-induced mouse model of DR. CONCLUSIONS In hyperglycemia, the TTR-induced increase in circ_0007411 could repress retinal neovascularization via the miR-548m/PTPN12/SKP1/EGFR pathway.
Collapse
Affiliation(s)
- Di Hu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Yikun Tian
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Lu Ye
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Yu Xin
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Jun Shao
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
9
|
Wei B, Zhu Y, Yang P, Han Y, Wang S, Wang X, Xia S, Song X, Zhang Z, Wang S, Rondard P, Pin JP, Jiang X, Liu J. GABA B1e promotes the malignancy of human cancer cells by targeting the tyrosine phosphatase PTPN12. iScience 2021; 24:103311. [PMID: 34778730 PMCID: PMC8577127 DOI: 10.1016/j.isci.2021.103311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/24/2021] [Accepted: 10/14/2021] [Indexed: 01/30/2023] Open
Abstract
Neurotransmitter receptors are involved in cancer progression. Among them, the heterodimeric GABAB receptor, activated by the main inhibitory neurotransmitter GABA, is composed of the transmembrane GABAB1 and GABAB2 subunits. The oncogenic role of the isoform GABAB1e (GB1e) containing only the extracellular domain of GABAB1 remains unclear. We revealed that GB1e is largely expressed in human breast cancer (BrCa) cell lines as well as in BrCa tissues where it is upregulated. Moreover, GB1e promoted the malignancy of BrCa cells both in vitro and in vivo. We propose that GB1e favors EGFR signaling by interacting with PTPN12 to disrupt the interaction between EGFR and PTPN12, and phosphorylation of Y230 and Y404 on GB1e is required in this process. Our data highlight that the GABBR1 gene through the expression of the GB1e isoform might play an important oncogenic role in BrCa and that GB1e is of interest for the treatment of some cancers. GABAB1e promotes the malignancy of breast cancer cells both in vitro and in vivo Specific phosphorylation of GABAB1e is critical for its association with PTPN12 GABAB1e disrupts EGFR interacting with PTPN12 and induces EGFR-PI3K/Akt signaling
Collapse
Affiliation(s)
- Bo Wei
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Yini Zhu
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Peng Yang
- Department of Breast & Endocrine Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China
| | - Yong Han
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan 430022, China
| | - Suyun Wang
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Xiaomei Wang
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Shuai Xia
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Xiaoguang Song
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Zhongling Zhang
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Sheng Wang
- Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Philippe Rondard
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Xinnong Jiang
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
- Corresponding author
| | - Jianfeng Liu
- Key Laboratory of Molecular Biophysics of MOE, International Research Center for Sensory Biology and Technology of MOST, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
- Corresponding author
| |
Collapse
|
10
|
Sarkar A, Kim EY, Jang T, Hongdusit A, Kim H, Choi JM, Fox JM. Microbially Guided Discovery and Biosynthesis of Biologically Active Natural Products. ACS Synth Biol 2021; 10:1505-1519. [PMID: 33988973 DOI: 10.1021/acssynbio.1c00074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The design of small molecules that inhibit disease-relevant proteins represents a longstanding challenge of medicinal chemistry. Here, we describe an approach for encoding this challenge-the inhibition of a human drug target-into a microbial host and using it to guide the discovery and biosynthesis of targeted, biologically active natural products. This approach identified two previously unknown terpenoid inhibitors of protein tyrosine phosphatase 1B (PTP1B), an elusive therapeutic target for the treatment of diabetes and cancer. Both inhibitors appear to target an allosteric site, which confers selectivity, and can inhibit PTP1B in living cells. A screen of 24 uncharacterized terpene synthases from a pool of 4464 genes uncovered additional hits, demonstrating a scalable discovery approach, and the incorporation of different PTPs into the microbial host yielded alternative PTP-specific detection systems. Findings illustrate the potential for using microbes to discover and build natural products that exhibit precisely defined biochemical activities yet possess unanticipated structures and/or binding sites.
Collapse
Affiliation(s)
- Ankur Sarkar
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Avenue, Boulder, Colorado 80303, United States
| | - Edward Y. Kim
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Avenue, Boulder, Colorado 80303, United States
| | - Taehwan Jang
- Department of Chemistry, Korea Advanced Institute of Science and Technology, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Akarawin Hongdusit
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Avenue, Boulder, Colorado 80303, United States
| | - Hyungjun Kim
- Department of Chemistry, Korea Advanced Institute of Science and Technology, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jeong-Mo Choi
- Department of Chemistry, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Jerome M. Fox
- Department of Chemical and Biological Engineering, University of Colorado Boulder, 3415 Colorado Avenue, Boulder, Colorado 80303, United States
| |
Collapse
|
11
|
Huang KL, Scott AD, Zhou DC, Wang LB, Weerasinghe A, Elmas A, Liu R, Wu Y, Wendl MC, Wyczalkowski MA, Baral J, Sengupta S, Lai CW, Ruggles K, Payne SH, Raphael B, Fenyö D, Chen K, Mills G, Ding L. Spatially interacting phosphorylation sites and mutations in cancer. Nat Commun 2021; 12:2313. [PMID: 33875650 PMCID: PMC8055881 DOI: 10.1038/s41467-021-22481-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 02/17/2021] [Indexed: 11/18/2022] Open
Abstract
Advances in mass-spectrometry have generated increasingly large-scale proteomics datasets containing tens of thousands of phosphorylation sites (phosphosites) that require prioritization. We develop a bioinformatics tool called HotPho and systematically discover 3D co-clustering of phosphosites and cancer mutations on protein structures. HotPho identifies 474 such hybrid clusters containing 1255 co-clustering phosphosites, including RET p.S904/Y928, the conserved HRAS/KRAS p.Y96, and IDH1 p.Y139/IDH2 p.Y179 that are adjacent to recurrent mutations on protein structures not found by linear proximity approaches. Hybrid clusters, enriched in histone and kinase domains, frequently include expression-associated mutations experimentally shown as activating and conferring genetic dependency. Approximately 300 co-clustering phosphosites are verified in patient samples of 5 cancer types or previously implicated in cancer, including CTNNB1 p.S29/Y30, EGFR p.S720, MAPK1 p.S142, and PTPN12 p.S275. In summary, systematic 3D clustering analysis highlights nearly 3,000 likely functional mutations and over 1000 cancer phosphosites for downstream investigation and evaluation of potential clinical relevance.
Collapse
Affiliation(s)
- Kuan-Lin Huang
- Department of Genetics and Genomics, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Adam D Scott
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Daniel Cui Zhou
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Liang-Bo Wang
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Amila Weerasinghe
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Abdulkadir Elmas
- Department of Genetics and Genomics, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruiyang Liu
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Yige Wu
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael C Wendl
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Jessika Baral
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Sohini Sengupta
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Chin-Wen Lai
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Kelly Ruggles
- Center for Health Informatics and Bioinformatics, New York University School of Medicine, New York, NY, USA
| | - Samuel H Payne
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Benjamin Raphael
- Lewis-Sigler Institute, Princeton University, Princeton, NJ, USA
| | - David Fenyö
- Center for Health Informatics and Bioinformatics, New York University School of Medicine, New York, NY, USA
| | - Ken Chen
- Departments of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gordon Mills
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Li Ding
- Department of Medicine, McDonnell Genome Institute, Department of Genetics, Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
12
|
Xu YF, Chen X, Yang Z, Xiao P, Liu CH, Li KS, Yang XZ, Wang YJ, Zhu ZL, Xu ZG, Zhang S, Wang C, Song YC, Zhao WD, Wang CH, Ji ZL, Zhang ZY, Cui M, Sun JP, Yu X. PTP-MEG2 regulates quantal size and fusion pore opening through two distinct structural bases and substrates. EMBO Rep 2021; 22:e52141. [PMID: 33764618 DOI: 10.15252/embr.202052141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/26/2021] [Accepted: 02/18/2021] [Indexed: 02/02/2023] Open
Abstract
Tyrosine phosphorylation of secretion machinery proteins is a crucial regulatory mechanism for exocytosis. However, the participation of protein tyrosine phosphatases (PTPs) in different exocytosis stages has not been defined. Here we demonstrate that PTP-MEG2 controls multiple steps of catecholamine secretion. Biochemical and crystallographic analyses reveal key residues that govern the interaction between PTP-MEG2 and its substrate, a peptide containing the phosphorylated NSF-pY83 site, specify PTP-MEG2 substrate selectivity, and modulate the fusion of catecholamine-containing vesicles. Unexpectedly, delineation of PTP-MEG2 mutants along with the NSF binding interface reveals that PTP-MEG2 controls the fusion pore opening through NSF independent mechanisms. Utilizing bioinformatics search and biochemical and electrochemical screening approaches, we uncover that PTP-MEG2 regulates the opening and extension of the fusion pore by dephosphorylating the DYNAMIN2-pY125 and MUNC18-1-pY145 sites. Further structural and biochemical analyses confirmed the interaction of PTP-MEG2 with MUNC18-1-pY145 or DYNAMIN2-pY125 through a distinct structural basis compared with that of the NSF-pY83 site. Our studies thus provide mechanistic insights in complex exocytosis processes.
Collapse
Affiliation(s)
- Yun-Fei Xu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China.,Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xu Chen
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Chun-Hua Liu
- Department of Physiology, Shandong First Medical University, Taian, China
| | - Kang-Shuai Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China.,Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao-Zhen Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yi-Jing Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Zhong-Liang Zhu
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zhi-Gang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, China
| | - Sheng Zhang
- Departments of Medicinal Chemistry and Molecular Pharmacology and of Chemistry, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Chuan Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - You-Chen Song
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, China Medical University, Shenyang, China
| | - Chang-He Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zhi-Liang Ji
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Zhong-Yin Zhang
- Departments of Medicinal Chemistry and Molecular Pharmacology and of Chemistry, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Min Cui
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, Jinan, China
| |
Collapse
|
13
|
Tong M, Liu P, Sun W, Liu J, Fan N, Wang X, Zhang Z, Song X, Lv C, Wang Y. Molecular dynamics simulation studies on the specific regulation of PTPN18 to the HER2 phospho-peptides. J Mol Recognit 2021; 34:e2890. [PMID: 33620127 DOI: 10.1002/jmr.2890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/15/2021] [Accepted: 01/26/2021] [Indexed: 11/09/2022]
Abstract
The specific regulation of PTPN18 protein to three HER2 phospho-peptides has been studied by molecular dynamics simulations and free energy calculations. The results revealed that the three HER2 phospho-peptides binding to the PTPN18 catalytic domain is energetically favorable due to substrate specificity of PTPN18, and moreover, the PTPN18 protein have significantly higher affinity to pY1248 peptide (-45.22 kcal/mol) than that of pY1112 (-25.3 kcal/mol) and pY1196 (-31.86 kcal/mol) peptides. Further, the binding of HER2 phospho-peptides to PTPN18 have also caused the closure of WPD-loop with the decrease of the centroid distances between the P-loop and the WPD loop. The WPD-loop closure of PTPN18 relates directly to the new hydrogen bond and hydrophobic interaction formations between the residues Tyr62, Asp64, Val65, Ala231, Arg235, and Ala273 in PTPN18 and Tyr(PO3) in the HER2 phospho-peptides, which suggests that these key residues would contribute to the specific regulation of PTPN18 to the substrates. The correlation analysis revealed the allosteric communication networks from the pY binding loop to the WPD loop through the structural change and the residue interactions in PTPN18. These results will be helpful to understand the specific regulation through the allosteric communication network in the PTPN18 catalytic domain.
Collapse
Affiliation(s)
- Mingqiong Tong
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou, China
| | - Peng Liu
- The Office of Academic Affairs, Dezhou University, Dezhou, China
| | - Wan Sun
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou, China
| | - Jing Liu
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou, China
| | - Na Fan
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou, China
| | - Xiaoyue Wang
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou, China
| | - Zhongyu Zhang
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou, China
| | - Xinfeng Song
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou, China
| | - Chao Lv
- Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, College of Medicine and Nursing, Dezhou University, Dezhou, China
| | - Yan Wang
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| |
Collapse
|
14
|
Chandel S, Manikandan A, Mehta N, Nathan AA, Tiwari RK, Mohapatra SB, Chandran M, Jaleel A, Manoj N, Dixit M. The protein tyrosine phosphatase PTP-PEST mediates hypoxia-induced endothelial autophagy and angiogenesis via AMPK activation. J Cell Sci 2021; 134:jcs250274. [PMID: 33323505 DOI: 10.1242/jcs.250274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/23/2020] [Indexed: 12/22/2022] Open
Abstract
Global and endothelial loss of PTP-PEST (also known as PTPN12) is associated with impaired cardiovascular development and embryonic lethality. Although hypoxia is implicated in vascular remodelling and angiogenesis, its effect on PTP-PEST remains unexplored. Here we report that hypoxia (1% oxygen) increases protein levels and catalytic activity of PTP-PEST in primary endothelial cells. Immunoprecipitation followed by mass spectrometry revealed that α subunits of AMPK (α1 and α2, encoded by PRKAA1 and PRKAA2, respectively) interact with PTP-PEST under normoxia but not in hypoxia. Co-immunoprecipitation experiments confirmed this observation and determined that AMPK α subunits interact with the catalytic domain of PTP-PEST. Knockdown of PTP-PEST abrogated hypoxia-mediated tyrosine dephosphorylation and activation of AMPK (Thr172 phosphorylation). Absence of PTP-PEST also blocked hypoxia-induced autophagy (LC3 degradation and puncta formation), which was rescued by the AMPK activator metformin (500 µM). Because endothelial autophagy is a prerequisite for angiogenesis, knockdown of PTP-PEST also attenuated endothelial cell migration and capillary tube formation, with autophagy inducer rapamycin (200 nM) rescuing angiogenesis. In conclusion, this work identifies for the first time that PTP-PEST is a regulator of hypoxia-induced AMPK activation and endothelial autophagy to promote angiogenesis.
Collapse
Affiliation(s)
- Shivam Chandel
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu 600036, India
| | - Amrutha Manikandan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu 600036, India
| | - Nikunj Mehta
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu 600036, India
| | - Abel Arul Nathan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu 600036, India
| | - Rakesh Kumar Tiwari
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu 600036, India
| | - Samar Bhallabha Mohapatra
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu 600036, India
| | - Mahesh Chandran
- Cardiovascular Disease and Diabetes Biology Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thyacaud Post, Thiruvananthpuram, Kerala 695014, India
| | - Abdul Jaleel
- Cardiovascular Disease and Diabetes Biology Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thyacaud Post, Thiruvananthpuram, Kerala 695014, India
| | - Narayanan Manoj
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu 600036, India
| | - Madhulika Dixit
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras (IIT Madras), Chennai, Tamil Nadu 600036, India
| |
Collapse
|
15
|
Huo YH, Wang YN, Meng LB, Zhang AL, Liu B. Progress in the correlation between PTPN12 gene expression and human tumors. Medicine (Baltimore) 2020; 99:e20445. [PMID: 32541467 PMCID: PMC7302617 DOI: 10.1097/md.0000000000020445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The global morbidity of cancer is rising rapidly. Despite advances in molecular biology, immunology, and cytotoxic and immune-anticancer therapies, cancer remains a major cause of death worldwide. Protein tyrosine phosphatase non-receptor type 12 (PTPN12) is a new member of the cytoplasmic protein tyrosine phosphatase family, isolated from a cDNA library of adult colon tissue. Thus far, no studies have reviewed the correlation between PTPN12 gene expression and human tumors. METHODS This article summarizes the latest domestic and international research developments on how the expression of PTPN12 relates to human tumors. The extensive search in Web of Science and PubMed with the keywords including PTPN12, tumor, renal cell carcinoma, proto-oncogenes, tumor suppressor genes was undertaken. RESULTS More and more studies have shown that a tumor is essentially a genetic disease, arising from a broken antagonistic function between proto-oncogenes and tumor suppressor genes. When their antagonistic effect is out of balance, it may cause uncontrolled growth of cells and lead to the occurrence of tumors. PTPN12 is a tumor suppressor gene, so inhibiting its activity will lead directly or indirectly to the occurrence of tumors. CONCLUSION The etiology, prevention, and treatment of tumors have become the focus of research around the world. PTPN12 is a tumor suppressor gene. In the future, PTPN12 might serve as a novel molecular marker to benefit patients, and even the development of tumor suppressor gene activation agents can form a practical research direction.
Collapse
Affiliation(s)
- Yu-hu Huo
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai
| | - Ya-ni Wang
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei
| | - Ling-bing Meng
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei
| | - Ai-li Zhang
- Department of Urinary Surgery, The fourth hospital of Hebei medical university, P. R. China
| | - Bin Liu
- Department of Urinary Surgery, The fourth hospital of Hebei medical university, P. R. China
| |
Collapse
|
16
|
Hou X, Sun JP, Ge L, Liang X, Li K, Zhang Y, Fang H. Inhibition of striatal-enriched protein tyrosine phosphatase by targeting computationally revealed cryptic pockets. Eur J Med Chem 2020; 190:112131. [PMID: 32078861 PMCID: PMC7163917 DOI: 10.1016/j.ejmech.2020.112131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 01/21/2020] [Accepted: 02/06/2020] [Indexed: 11/21/2022]
Abstract
Cryptic pockets, which are not apparent in crystallographic structures, provide promising alternatives to traditional binding sites for drug development. However, identifying cryptic pockets is extremely challenging and the therapeutic potential of cryptic pockets remains unclear. Here, we reported the discovery of novel inhibitors for striatal-enriched protein tyrosine phosphatase (STEP), a potential drug target for multiple neuropsychiatric disorders, based on cryptic pocket detection. By combining the use of molecular dynamics simulations and fragment-centric topographical mapping, we identified transiently open cryptic pockets and identified 12 new STEP inhibition scaffolds through structure-based virtual screening. Site-directed mutagenesis verified the binding of ST3 with the predicted cryptic pockets. Moreover, the most potent and selective inhibitors could modulate the phosphorylation of both ERK1/2 and Pyk2 in PC12 cells.
Collapse
Affiliation(s)
- Xuben Hou
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmaceutical Science, Shandong University, Jinan, Shandong, 250012, China; Department of Chemistry, New York University, New York, NY, 10003, United States
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lin Ge
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xiao Liang
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmaceutical Science, Shandong University, Jinan, Shandong, 250012, China
| | - Kangshuai Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yingkai Zhang
- Department of Chemistry, New York University, New York, NY, 10003, United States; NYU-ECNU Center for Computational Chemistry, New York University-Shanghai, Shanghai, 200122, China
| | - Hao Fang
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmaceutical Science, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
17
|
Weidemann SA, Sauer C, Luebke AM, Möller-Koop C, Steurer S, Hube-Magg C, Büscheck F, Höflmayer D, Tsourlakis MC, Clauditz TS, Simon R, Sauter G, Göbel C, Lebok P, Dum D, Fraune C, Kind S, Minner S, Izbicki J, Schlomm T, Huland H, Heinzer H, Burandt E, Haese A, Graefen M, Heumann A. High-level expression of protein tyrosine phosphatase non-receptor 12 is a strong and independent predictor of poor prognosis in prostate cancer. BMC Cancer 2019; 19:944. [PMID: 31606028 PMCID: PMC6790047 DOI: 10.1186/s12885-019-6182-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/20/2019] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Protein tyrosine phosphatase non-receptor 12 (PTPN12) is ubiquitously tyrosine phosphatase with tumor suppressive properties. METHODS PTPN12 expression was analyzed by immunohistochemistry on a tissue microarray with 13,660 clinical prostate cancer specimens. RESULTS PTPN12 staining was typically absent or weak in normal prostatic epithelium but seen in the majority of cancers, where staining was considered weak in 26.5%, moderate in 39.9%, and strong in 4.7%. High PTPN12 staining was associated with high pT category, high classical and quantitative Gleason grade, lymph node metastasis, positive surgical margin, high Ki67 labeling index and early prostate specific antigen recurrence (p < 0.0001 each). PTPN12 staining was seen in 86.4% of TMPRSS2:ERG fusion positive but in only 58.4% of ERG negative cancers. Subset analyses discovered that all associations with unfavorable phenotype and prognosis were markedly stronger in ERG positive than in ERG negative cancers but still retained in the latter group. Multivariate analyses revealed an independent prognostic impact of high PTPN12 expression in all cancers and in the ERG negative subgroup and to a lesser extent also in ERG positive cancers. Comparison with 12 previously analyzed chromosomal deletions revealed that high PTPN12 expression was significantly associated with 10 of 12 deletions in ERG negative and with 7 of 12 deletions in ERG positive cancers (p < 0.05 each) indicating that PTPN12 overexpression parallels increased genomic instability in prostate cancer. CONCLUSIONS These data identify PTPN12 as an independent prognostic marker in prostate cancer. PTPN12 analysis, either alone or in combination with other biomarkers might be of clinical utility in assessing prostate cancer aggressiveness.
Collapse
Affiliation(s)
- Sören A Weidemann
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Charlotte Sauer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Andreas M Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Christina Möller-Koop
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Doris Höflmayer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Maria Christina Tsourlakis
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Cosima Göbel
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Simon Kind
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Jakob Izbicki
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Thorsten Schlomm
- Department of Urology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Hartwig Huland
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg, Eppendorf, Germany
| | - Hans Heinzer
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg, Eppendorf, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Alexander Haese
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg, Eppendorf, Germany
| | - Markus Graefen
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg, Eppendorf, Germany
| | - Asmus Heumann
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| |
Collapse
|
18
|
Lee C, Rhee I. Important roles of protein tyrosine phosphatase PTPN12 in tumor progression. Pharmacol Res 2019; 144:73-78. [DOI: 10.1016/j.phrs.2019.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 03/26/2019] [Accepted: 04/04/2019] [Indexed: 12/27/2022]
|
19
|
Dolatabad MR, Guo LL, Xiao P, Zhu Z, He QT, Yang DX, Qu CX, Guo SC, Fu XL, Li RR, Ge L, Hu KJ, Liu HD, Shen YM, Yu X, Sun JP, Zhang PJ. Crystal structure and catalytic activity of the PPM1K N94K mutant. J Neurochem 2019; 148:550-560. [DOI: 10.1111/jnc.14631] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Meisam Rostaminasab Dolatabad
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| | - Lu-lu Guo
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
- Key Laboratory of Chemical Biology; Ministry of Education; Shandong University School of Pharmaceutical Science; Jinan Shandong China
| | - Zhongliang Zhu
- School of Life Sciences; University of Science and Technology of China; Hefei Anhui China
| | - Qing-tao He
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| | - Du-xiao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| | - Chang-xiu Qu
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| | - Sheng-chao Guo
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| | - Xiao-lei Fu
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| | - Rui-rui Li
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| | - Lin Ge
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| | - Ke-jia Hu
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| | - Hong-da Liu
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
- Department of Pharmacology and Chemical Biology; School of Medicine; University of Pittsburgh; Pittsburgh Pennsylvania USA
| | - Yue-mao Shen
- Key Laboratory of Chemical Biology; Ministry of Education; Shandong University School of Pharmaceutical Science; Jinan Shandong China
| | - Xiao Yu
- Department of Physiology; Shandong University; School of Medicine; Jinan Shandong China
| | - Jin-peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
- Department of Physiology and Pathophysiology; School of Basic Medical Sciences; Peking University; Key Laboratory of Molecular Cardiovascular Science; Ministry of Education; Beijing China
| | - Peng-ju Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education; Department of Biochemistry and Molecular Biology; Shandong University School of Medicine; Jinan Shandong China
| |
Collapse
|
20
|
Akter S, Fu L, Jung Y, Conte ML, Lawson JR, Lowther WT, Sun R, Liu K, Yang J, Carroll KS. Chemical proteomics reveals new targets of cysteine sulfinic acid reductase. Nat Chem Biol 2018; 14:995-1004. [PMID: 30177848 PMCID: PMC6192846 DOI: 10.1038/s41589-018-0116-2] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 07/18/2018] [Indexed: 01/06/2023]
Abstract
Cysteine sulfinic acid or S-sulfinylation is an oxidative post-translational modification (OxiPTM) that is known to be involved in redox-dependent regulation of protein function but has been historically difficult to analyze biochemically. To facilitate the detection of S-sulfinylated proteins, we demonstrate that a clickable, electrophilic diazene probe (DiaAlk) enables capture and site-centric proteomic analysis of this OxiPTM. Using this workflow, we revealed a striking difference between sulfenic acid modification (S-sulfenylation) and the S-sulfinylation dynamic response to oxidative stress, which is indicative of different roles for these OxiPTMs in redox regulation. We also identified >55 heretofore-unknown protein substrates of the cysteine sulfinic acid reductase sulfiredoxin, extending its function well beyond those of 2-cysteine peroxiredoxins (2-Cys PRDX1-4) and offering new insights into the role of this unique oxidoreductase as a central mediator of reactive oxygen species-associated diseases, particularly cancer. DiaAlk therefore provides a novel tool to profile S-sulfinylated proteins and study their regulatory mechanisms in cells.
Collapse
Affiliation(s)
- Salma Akter
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA
| | - Ling Fu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing, China
- National Center for Protein Sciences-Beijing, Beijing, China
- Beijing Institute of Lifeomics, Beijing, China
| | - Youngeun Jung
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA
| | - Mauro Lo Conte
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA
- Novo Nordisk Research Center, Seattle, WA, USA
| | - J Reed Lawson
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - W Todd Lowther
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Structural Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Rui Sun
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing, China
- National Center for Protein Sciences-Beijing, Beijing, China
- Beijing Institute of Lifeomics, Beijing, China
| | - Keke Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing, China
- National Center for Protein Sciences-Beijing, Beijing, China
- Beijing Institute of Lifeomics, Beijing, China
| | - Jing Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing, China.
- National Center for Protein Sciences-Beijing, Beijing, China.
- Beijing Institute of Lifeomics, Beijing, China.
| | - Kate S Carroll
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA.
| |
Collapse
|
21
|
Tang H, Dai Z, Qin X, Cai W, Hu L, Huang Y, Cao W, Yang F, Wang C, Liu T. Proteomic Identification of Protein Tyrosine Phosphatase and Substrate Interactions in Living Mammalian Cells by Genetic Encoding of Irreversible Enzyme Inhibitors. J Am Chem Soc 2018; 140:13253-13259. [PMID: 30247891 DOI: 10.1021/jacs.8b06922] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Protein tyrosine phosphatases (PTPs) play critical roles in cell signaling pathways, but identification of unknown PTPs for a given substrate in live cells remain technically challenging. Here, we synthesized a series of tyrosine-based irreversible PTP inhibitors and characterized by site-specific encoding on substrate proteins in cells with an expanded genetic code. By fine-tuning the chemical reactivity, we identified optimal active amino acid probes to covalently cross-link a PTP and its substrate both in vitro and in mammalian cells. Using HER2 as an example, we provide first direct evidence of HER2 Y1023 and SHP2 cross-linking in situ in living human cells. Moreover, proteomic analysis using our approach identified PTP1B as a novel phosphatase for HER2 that specifically dephosphorylated pY1221 position, which may shed light on the puzzle of PTP1B's role in HER2 positive breast cancer. This novel method provides a useful tool for dissecting tyrosine phosphoregulation in living cells.
Collapse
Affiliation(s)
- Hongting Tang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences , Peking University , 38 Xueyuan Road , Haidian District, Beijing 100191 , China
| | - Zhen Dai
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences , Peking University , 38 Xueyuan Road , Haidian District, Beijing 100191 , China.,College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry , Nankai University , Tianjin 300071 , China
| | - Xuewen Qin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences , Peking University , 38 Xueyuan Road , Haidian District, Beijing 100191 , China
| | - Wenkang Cai
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences , Peking University , 38 Xueyuan Road , Haidian District, Beijing 100191 , China
| | - Liming Hu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences , Peking University , 38 Xueyuan Road , Haidian District, Beijing 100191 , China
| | - Yujia Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences , Peking University , 38 Xueyuan Road , Haidian District, Beijing 100191 , China
| | - Wenbing Cao
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences , Peking University , 38 Xueyuan Road , Haidian District, Beijing 100191 , China.,College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry , Nankai University , Tianjin 300071 , China
| | - Fan Yang
- College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , China
| | - Chu Wang
- College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , China
| | - Tao Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences , Peking University , 38 Xueyuan Road , Haidian District, Beijing 100191 , China
| |
Collapse
|
22
|
Yang D, Xiao P, Li Q, Fu X, Pan C, Lu D, Wen S, Xia W, He D, Li H, Fang H, Shen Y, Xu Z, Lin A, Wang C, Yu X, Wu J, Sun J. Allosteric modulation of the catalytic VYD loop in Slingshot by its N-terminal domain underlies both Slingshot auto-inhibition and activation. J Biol Chem 2018; 293:16226-16241. [PMID: 30154244 DOI: 10.1074/jbc.ra118.004175] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/21/2018] [Indexed: 12/22/2022] Open
Abstract
Slingshots are phosphatases that modulate cytoskeleton dynamics, and their activities are tightly regulated in different physiological contexts. Recently, abnormally elevated Slingshot activity has been implicated in many human diseases, such as cancer, Alzheimer's disease, and vascular diseases. Therefore, Slingshot-specific inhibitors have therapeutic potential. However, an enzymological understanding of the catalytic mechanism of Slingshots and of their activation by actin is lacking. Here, we report that the N-terminal region of human Slingshot2 auto-inhibits its phosphatase activity in a noncompetitive manner. pH-dependent phosphatase assays and leaving-group dependence studies suggested that the N-terminal domain of Slingshot2 regulates the stability of the leaving group of the product during catalysis by modulating the general acid Asp361 in the catalytic VYD loop. F-actin binding relieved this auto-inhibition and restored the function of the general acid. Limited tryptic digestion and biophysical studies identified large conformational changes in Slingshot2 after the F-actin binding. The dissociation of N-terminal structural elements, including Leu63, and the exposure of the loop between α-helix-2 and β-sheet-3 of the phosphatase domain served as the structural basis for Slingshot activation via F-actin binding in vitro and via neuregulin stimulation in cells. Moreover, we designed a FlAsH-BRET-based Slingshot2 biosensor whose readout was highly correlated with the in vivo phosphatase activities of Slingshot2. Our results reveal the auto-inhibitory mechanism and allosteric activation mechanisms of a human Slingshot phosphatase. They also contribute to the design of new strategies to study Slingshot regulation in various cellular contexts and to screen for new activators/inhibitors of Slingshot activity.
Collapse
Affiliation(s)
- Duxiao Yang
- From the Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, and
| | - Peng Xiao
- From the Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, and.,the School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Qing Li
- the Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Xiaolei Fu
- From the Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, and
| | - Chang Pan
- From the Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, and
| | - Di Lu
- From the Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, and
| | - Shishuai Wen
- the School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Wanying Xia
- From the Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, and
| | - Dongfang He
- From the Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, and
| | - Hui Li
- the Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Hao Fang
- the School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yuemao Shen
- the School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Zhigang Xu
- the School of Life Science, Shandong University, Jinan, Shandong 250003, China
| | - Amy Lin
- the School of Medicine, Duke University, Durham, North Carolina 27705
| | - Chuan Wang
- the Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiao Yu
- the Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Jiawei Wu
- the MOE Key Laboratory of Protein Science, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jinpeng Sun
- From the Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, and .,the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China, and.,the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing 100191, China
| |
Collapse
|
23
|
Yang F, Xiao P, Qu CX, Liu Q, Wang LY, Liu ZX, He QT, Liu C, Xu JY, Li RR, Li MJ, Li Q, Guo XZ, Yang ZY, He DF, Yi F, Ruan K, Shen YM, Yu X, Sun JP, Wang J. Allosteric mechanisms underlie GPCR signaling to SH3-domain proteins through arrestin. Nat Chem Biol 2018; 14:876-886. [PMID: 30120361 DOI: 10.1038/s41589-018-0115-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 07/03/2018] [Indexed: 12/30/2022]
Abstract
Signals from 800 G-protein-coupled receptors (GPCRs) to many SH3 domain-containing proteins (SH3-CPs) regulate important physiological functions. These GPCRs may share a common pathway by signaling to SH3-CPs via agonist-dependent arrestin recruitment rather than through direct interactions. In the present study, 19F-NMR and cellular studies revealed that downstream of GPCR activation engagement of the receptor-phospho-tail with arrestin allosterically regulates the specific conformational states and functional outcomes of remote β-arrestin 1 proline regions (PRs). The observed NMR chemical shifts of arrestin PRs were consistent with the intrinsic efficacy and specificity of SH3 domain recruitment, which was controlled by defined propagation pathways. Moreover, in vitro reconstitution experiments and biophysical results showed that the receptor-arrestin complex promoted SRC kinase activity through an allosteric mechanism. Thus, allosteric regulation of the conformational states of β-arrestin 1 PRs by GPCRs and the allosteric activation of downstream effectors by arrestin are two important mechanisms underlying GPCR-to-SH3-CP signaling.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China.,Institute of Biophysics, Chinese Academy of Sciences, Beijing, Chaoyang district, Beijing, China.,Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, Shandong, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China.,Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Science, Shandong University, Jinan, Shandong, China
| | - Chang-Xiu Qu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Qi Liu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China.,Institute of Biophysics, Chinese Academy of Sciences, Beijing, Chaoyang district, Beijing, China.,Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, Shandong, China
| | - Liu-Yang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA
| | - Zhi-Xin Liu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Qing-Tao He
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Chuan Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Jian-Ye Xu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Rui-Rui Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Meng-Jing Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Qing Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, Shandong, China
| | - Xu-Zhen Guo
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, Chaoyang district, Beijing, China
| | - Zhao-Ya Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Dong-Fang He
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Ke Ruan
- Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei Anhui, China
| | - Yue-Mao Shen
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Medicine, Shandong, China
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China. .,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.
| | - Jiangyun Wang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, Chaoyang district, Beijing, China.
| |
Collapse
|
24
|
Chen Z, Morales JE, Guerrero PA, Sun H, McCarty JH. PTPN12/PTP-PEST Regulates Phosphorylation-Dependent Ubiquitination and Stability of Focal Adhesion Substrates in Invasive Glioblastoma Cells. Cancer Res 2018; 78:3809-3822. [PMID: 29743287 DOI: 10.1158/0008-5472.can-18-0085] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/03/2018] [Accepted: 05/07/2018] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is an invasive brain cancer with tumor cells that disperse from the primary mass, escaping surgical resection and invariably giving rise to lethal recurrent lesions. Here we report that PTP-PEST, a cytoplasmic protein tyrosine phosphatase, controls GBM cell invasion by physically bridging the focal adhesion protein Crk-associated substrate (Cas) to valosin-containing protein (Vcp), an ATP-dependent protein segregase that selectively extracts ubiquitinated proteins from multiprotein complexes and targets them for degradation via the ubiquitin proteasome system. Both Cas and Vcp are substrates for PTP-PEST, with the phosphorylation status of tyrosine 805 (Y805) in Vcp impacting affinity for Cas in focal adhesions and controlling ubiquitination levels and protein stability. Perturbing PTP-PEST-mediated phosphorylation of Cas and Vcp led to alterations in GBM cell-invasive growth in vitro and in preclinical mouse models. Collectively, these data reveal a novel regulatory mechanism involving PTP-PEST, Vcp, and Cas that dynamically balances phosphorylation-dependent ubiquitination of key focal proteins involved in GBM cell invasion.Significance: PTP-PEST balances GBM cell growth and invasion by interacting with the ATP-dependent ubiquitin segregase Vcp/p97 and regulating phosphorylation and stability of the focal adhesion protein p130Cas.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/14/3809/F1.large.jpg Cancer Res; 78(14); 3809-22. ©2018 AACR.
Collapse
Affiliation(s)
- Zhihua Chen
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John E Morales
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paola A Guerrero
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Huandong Sun
- Institute for Applied Cancer Sciences, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph H McCarty
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
25
|
Combinatorial inhibition of PTPN12-regulated receptors leads to a broadly effective therapeutic strategy in triple-negative breast cancer. Nat Med 2018; 24:505-511. [PMID: 29578538 DOI: 10.1038/nm.4507] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/29/2018] [Indexed: 12/28/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer diagnosed in more than 200,000 women each year and is recalcitrant to targeted therapies. Although TNBCs harbor multiple hyperactive receptor tyrosine kinases (RTKs), RTK inhibitors have been largely ineffective in TNBC patients thus far. We developed a broadly effective therapeutic strategy for TNBC that is based on combined inhibition of receptors that share the negative regulator PTPN12. Previously, we and others identified the tyrosine phosphatase PTPN12 as a tumor suppressor that is frequently inactivated in TNBC. PTPN12 restrains several RTKs, suggesting that PTPN12 deficiency leads to aberrant activation of multiple RTKs and a co-dependency on these receptors. This in turn leads to the therapeutic hypothesis that PTPN12-deficient TNBCs may be responsive to combined RTK inhibition. However, the repertoire of RTKs that are restrained by PTPN12 in human cells has not been systematically explored. By methodically identifying the suite of RTK substrates (MET, PDGFRβ, EGFR, and others) inhibited by PTPN12, we rationalized a combination RTK-inhibitor therapy that induced potent tumor regression across heterogeneous models of TNBC. Orthogonal approaches revealed that PTPN12 was recruited to and inhibited these receptors after ligand stimulation, thereby serving as a feedback mechanism to limit receptor signaling. Cancer-associated mutation of PTPN12 or reduced PTPN12 protein levels diminished this feedback mechanism, leading to aberrant activity of these receptors. Restoring PTPN12 protein levels restrained signaling from RTKs, including PDGFRβ and MET, and impaired TNBC survival. In contrast with single agents, combined inhibitors targeting the PDGFRβ and MET receptors induced the apoptosis in TNBC cells in vitro and in vivo. This therapeutic strategy resulted in tumor regressions in chemo-refractory patient-derived TNBC models. Notably, response correlated with PTPN12 deficiency, suggesting that impaired receptor feedback may establish a combined addiction to these proto-oncogenic receptors. Taken together, our data provide a rationale for combining RTK inhibitors in TNBC and other malignancies that lack receptor-activating mutations.
Collapse
|
26
|
Proteomic Identification of Heat Shock-Induced Danger Signals in a Melanoma Cell Lysate Used in Dendritic Cell-Based Cancer Immunotherapy. J Immunol Res 2018; 2018:3982942. [PMID: 29744371 PMCID: PMC5878886 DOI: 10.1155/2018/3982942] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/28/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022] Open
Abstract
Autologous dendritic cells (DCs) loaded with cancer cell-derived lysates have become a promising tool in cancer immunotherapy. During the last decade, we demonstrated that vaccination of advanced melanoma patients with autologous tumor antigen presenting cells (TAPCells) loaded with an allogeneic heat shock- (HS-) conditioned melanoma cell-derived lysate (called TRIMEL) is able to induce an antitumor immune response associated with a prolonged patient survival. TRIMEL provides not only a broad spectrum of potential melanoma-associated antigens but also danger signals that are crucial in the induction of a committed mature DC phenotype. However, potential changes induced by heat conditioning on the proteome of TRIMEL are still unknown. The identification of newly or differentially expressed proteins under defined stress conditions is relevant for understanding the lysate immunogenicity. Here, we characterized the proteomic profile of TRIMEL in response to HS treatment. A quantitative label-free proteome analysis of over 2800 proteins was performed, with 91 proteins that were found to be regulated by HS treatment: 18 proteins were overexpressed and 73 underexpressed. Additionally, 32 proteins were only identified in the HS-treated TRIMEL and 26 in non HS-conditioned samples. One protein from the overexpressed group and two proteins from the HS-exclusive group were previously described as potential damage-associated molecular patterns (DAMPs). Some of the HS-induced proteins, such as haptoglobin, could be also considered as DAMPs and candidates for further immunological analysis in the establishment of new putative danger signals with immunostimulatory functions.
Collapse
|
27
|
Lin Q, Wang H, Lin X, Zhang W, Huang S, Zheng Y. PTPN12 Affects Nasopharyngeal Carcinoma Cell Proliferation and Migration Through Regulating EGFR. Cancer Biother Radiopharm 2018; 33:60-64. [PMID: 29634414 DOI: 10.1089/cbr.2017.2254] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Nasopharyngeal carcinoma (NPC) shows the leading morbidity in otorhinolaryngological malignant tumor. It is a common malignancy in China with obvious reginal distribution. NPC is a polygenic disease that is affected by numerous factors. Protein tyrosine phosphatase nonreceptor type 12 (PTPN12) regulates multiple tumor proliferation and development, including breast cancer and colon cancer. However, the role of PTPN12 in NPC occurrence and development has not been elucidated. PATIENTS AND METHODS NPC cell line CNE2 was cultured in vitro and divided into three groups, including control, empty plasmid, and PTPN12 groups. PTPN12 mRNA and protein expressions were tested by real-time polymerase chain reaction and Western blot. CNE2 cell proliferation was detected by MTT assay. Cell migration was determined by wound healing assay. Cell apoptosis was evaluated by caspase 3 activity detection. Epidermal growth factor receptor (EGFR) expression was assessed by Western blot. RESULTS PTPN12 plasmid transfection increased PTPN12 mRNA and protein expressions, suppressed cell proliferation and migration, reduced EGFR level, and enhanced caspase 3 activity compared with control and empty plasmid groups (p < 0.05). CONCLUSIONS PTPN12 regulates NPC proliferation and migration through negative regulating EGFR. It could be treated as a molecular target for NPC diagnosis and prognosis analysis.
Collapse
Affiliation(s)
- Qinghai Lin
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| | - Huige Wang
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| | - Xinqiang Lin
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| | - Wenrui Zhang
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| | - Shuhua Huang
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| | - Yandan Zheng
- Department of Otolaryngology, First Affiliated Hospital of Shantou University Medical College , Shantou, Guangdong, China
| |
Collapse
|
28
|
Clearance of a persistent picornavirus infection is associated with enhanced pro-apoptotic and cellular immune responses. Sci Rep 2017; 7:17800. [PMID: 29259271 PMCID: PMC5736604 DOI: 10.1038/s41598-017-18112-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 12/04/2017] [Indexed: 12/22/2022] Open
Abstract
Long-term persistent viral infections cause substantial morbidity and associated economic losses in human and veterinary contexts. Yet, the mechanisms associated with establishment of persistent infections are poorly elucidated. We investigated immunomodulatory mechanisms associated with clearance versus persistence of foot-and-mouth disease virus (FMDV) in micro-dissected compartments of the bovine nasopharynx by microarray. The use of laser-capture microdissection allowed elucidation of differential gene regulation within distinct anatomic compartments critical to FMDV infection. Analysis of samples from transitional and persistent phases of infection demonstrated significant differences in transcriptome profiles of animals that cleared infection versus those that became persistently infected carriers. Specifically, it was demonstrated that clearance of FMDV from the nasopharyngeal mucosa was associated with upregulation of targets associated with activation of T cell-mediated immunity. Contrastingly, gene regulation in FMDV carriers suggested inhibition of T cell activation and promotion of Th2 polarization. These findings were corroborated by immunofluorescence microscopy which demonstrated relative abundance of CD8+ T cells in the nasopharyngeal mucosa in association with clearance of FMDV. The findings presented herein emphasize that a critical balance between Th1 and Th2 -mediated immunity is essential for successful clearance of FMDV infection and should be considered for development of next-generation vaccines and antiviral products.
Collapse
|
29
|
Sui Y, Fu X, Wang Y, Hu W, Zhang T, Liu W, Jiang L, Xing S, Fu X, Xu X. Expression, purification and characterization of a catalytic domain of human protein tyrosine phosphatase non-receptor 12 (PTPN12) in Escherichia coli with FKBP-type PPIase as a chaperon. Protein Expr Purif 2017; 142:45-52. [PMID: 28965803 DOI: 10.1016/j.pep.2017.09.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/25/2017] [Accepted: 09/21/2017] [Indexed: 12/11/2022]
Abstract
Protein tyrosine phosphatase non-receptor type 12 (PTPN12), also known as PTP-PEST, was broadly expressed in hemopoietic cells. Recent research has shown that this enzyme is involved in tumorigenesis, as well as in tumor progression and transfer, as it can suppress multiple oncogenic tyrosine kinases. However, the difficulty of soluble expression of PTP-PEST in prokaryotic cells has resulted in great limitations in investigating its structure and functions. In this study, we successfully carried out soluble expression of the catalytic domain of PTP-PEST (ΔPTP-PEST) in Escherichia coli and performed an enzymatic characterization and kinetics. To confirm expression efficiency, we also induced the expression of the chaperon, FKBP_C. FKBP_C expression indicated efficacious prokaryotic expression of ΔPTP-PEST. In conclusion, our work yielded a practical expression system and two-step chromatography purification method that may serve as a valuable tool for the structural and functional analysis of proteins that are difficult to express in the soluble form in prokaryotic cells.
Collapse
Affiliation(s)
- Yuan Sui
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Xingye Fu
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Yuchen Wang
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Weiyan Hu
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Tong Zhang
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Wanyao Liu
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Liyan Jiang
- Core Facilities for Life Science, Jilin University, Changchun 130012, PR China
| | - Shu Xing
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun 130012, PR China.
| | - Xueqi Fu
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun 130012, PR China.
| | - Xuesong Xu
- Clinical Laboratory of China-Japan Union Hospital, Jilin University, Changchun 130033, PR China.
| |
Collapse
|
30
|
Li H, Yang D, Ning S, Xu Y, Yang F, Yin R, Feng T, Han S, Guo L, Zhang P, Qu W, Guo R, Song C, Xiao P, Zhou C, Xu Z, Sun J, Yu X. Switching of the substrate specificity of protein tyrosine phosphatase N12 by cyclin‐dependent kinase 2 phosphorylation orchestrating 2 oncogenic pathways. FASEB J 2017; 32:73-82. [PMID: 28842430 DOI: 10.1096/fj.201700418r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/14/2017] [Indexed: 01/27/2023]
Affiliation(s)
- Hui Li
- Key Laboratory Experimental Teratology of the Ministry of EducationShangdong University Jinan China
- Department of PhysiologyShangdong University Jinan China
- Second Hospital, Shangdong University Jinan China
| | - Duxiao Yang
- Department of Molecular Biology and BiochemistryShandong University School of Medicine, Shangdong University Jinan China
| | - Shanglei Ning
- Qilu Hospital and School of Life Science, Shangdong University Jinan China
| | - Yinghui Xu
- Cancer CenterFirst Hospital of Jilin University Jilin China
| | - Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of EducationShangdong University Jinan China
- Department of PhysiologyShangdong University Jinan China
| | - Rusha Yin
- Key Laboratory Experimental Teratology of the Ministry of EducationShangdong University Jinan China
- Department of PhysiologyShangdong University Jinan China
| | - Taihu Feng
- Key Laboratory Experimental Teratology of the Ministry of EducationShangdong University Jinan China
- Department of PhysiologyShangdong University Jinan China
| | - Shouqing Han
- Key Laboratory Experimental Teratology of the Ministry of EducationShangdong University Jinan China
- Department of PhysiologyShangdong University Jinan China
| | - Lu Guo
- Second Hospital, Shangdong University Jinan China
| | - Pengju Zhang
- Department of Molecular Biology and BiochemistryShandong University School of Medicine, Shangdong University Jinan China
| | - Wenjie Qu
- Key Laboratory Experimental Teratology of the Ministry of EducationShangdong University Jinan China
- Department of PhysiologyShangdong University Jinan China
| | - Renbo Guo
- Key Laboratory Experimental Teratology of the Ministry of EducationShangdong University Jinan China
- Department of PhysiologyShangdong University Jinan China
| | - Chen Song
- Center for Quantitative BiologyPeking University Beijing China
| | - Peng Xiao
- Department of Molecular Biology and BiochemistryShandong University School of Medicine, Shangdong University Jinan China
| | | | - Zhigang Xu
- Key Laboratory Experimental Teratology of the Ministry of EducationShangdong University Jinan China
| | - Jin‐Peng Sun
- Department of Molecular Biology and BiochemistryShandong University School of Medicine, Shangdong University Jinan China
- Duke University School of Medicine, Duke University Durham North Carolina USA
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of EducationShangdong University Jinan China
- Department of PhysiologyShangdong University Jinan China
| |
Collapse
|