1
|
Qian Z, Song D, Ipsaro JJ, Bautista C, Joshua-Tor L, Yeh JTH, Tonks NK. Manipulating PTPRD function with ectodomain antibodies. Genes Dev 2023; 37:743-759. [PMID: 37669874 PMCID: PMC10546974 DOI: 10.1101/gad.350713.123] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/28/2023] [Indexed: 09/07/2023]
Abstract
Protein tyrosine phosphatases (PTPs) are critical regulators of signal transduction but have yet to be exploited fully for drug development. Receptor protein tyrosine phosphatase δ (RPTPδ/PTPRD) has been shown to elicit tumor-promoting functions, including elevating SRC activity and promoting metastasis in certain cell contexts. Dimerization has been implicated in the inhibition of receptor protein tyrosine phosphatases (RPTPs). We have generated antibodies targeting PTPRD ectodomains with the goal of manipulating their dimerization status ectopically, thereby regulating intracellular signaling. We have validated antibody binding to endogenous PTPRD in a metastatic breast cancer cell line, CAL51, and demonstrated that a monoclonal antibody, RD-43, inhibited phosphatase activity and induced the degradation of PTPRD. Similar effects were observed following chemically induced dimerization of its phosphatase domain. Mechanistically, RD-43 triggered the formation of PTPRD dimers in which the phosphatase activity was impaired. Subsequently, the mAb-PTPRD dimer complex was degraded through lysosomal and proteasomal pathways, independently of secretase cleavage. Consequently, treatment with RD-43 inhibited SRC signaling and suppressed PTPRD-dependent cell invasion. Together, these findings demonstrate that manipulating RPTP function via antibodies to the extracellular segments has therapeutic potential.
Collapse
Affiliation(s)
- Zhe Qian
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- Graduate Program of Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York 11760, USA
| | - Dongyan Song
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Jonathan J Ipsaro
- Howard Hughes Medical Institute, W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | | | - Leemor Joshua-Tor
- Howard Hughes Medical Institute, W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Johannes T-H Yeh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Nicholas K Tonks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
| |
Collapse
|
2
|
Wu L, Zhou L, An J, Shao X, Zhang H, Wang C, Zhao G, Chen S, Cui X, Zhang X, Yang F, Li X, Zhang X. Comprehensive profiling of extracellular vesicles in uveitis and scleritis enables biomarker discovery and mechanism exploration. J Transl Med 2023; 21:388. [PMID: 37322475 PMCID: PMC10273650 DOI: 10.1186/s12967-023-04228-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Uveitis and posterior scleritis are sight-threatening diseases with undefined pathogenesis and accurate diagnosis remains challenging. METHODS Two plasma-derived extracellular vesicle (EV) subpopulations, small and large EVs, obtained from patients with ankylosing spondylitis-related uveitis, Behcet's disease uveitis, Vogt-Koyanagi-Harada syndrome, and posterior scleritis were subjected to proteomics analysis alongside plasma using SWATH-MS. A comprehensive bioinformatics analysis was performed on the proteomic profiles of sEVs, lEVs, and plasma. Candidate biomarkers were validated in a new cohort using ELISA. Pearson correlation analysis was performed to analyze the relationship between clinical parameters and proteomic data. Connectivity map database was used to predict therapeutic agents. RESULTS In total, 3,668 proteins were identified and over 3000 proteins were quantified from 278 samples. When comparing diseased group to healthy control, the proteomic profiles of the two EV subgroups were more correlated with disease than plasma. Comprehensive bioinformatics analysis highlighted potential pathogenic mechanisms for these diseases. Potential biomarker panels for four diseases were identified and validated. We found a negative correlation between plasma endothelin-converting enzyme 1 level and mean retinal thickness. Potential therapeutic drugs were proposed, and their targets were identified. CONCLUSIONS This study provides a proteomic landscape of plasma and EVs involved in ankylosing spondylitis-related uveitis, Behcet's disease uveitis, Vogt-Koyanagi-Harada syndrome, and posterior scleritis, offers insights into disease pathogenesis, identifies valuable biomarker candidates, and proposes promising therapeutic agents.
Collapse
Affiliation(s)
- Lingzi Wu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Lei Zhou
- Department of Applied Biology and Chemical Technology, School of Optometry, Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong, China
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, China
| | - Jinying An
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Xianfeng Shao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Hui Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Chunxi Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | | | - Shuang Chen
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Xuexue Cui
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Xinyi Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Fuhua Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Xiaomin Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China.
| |
Collapse
|
3
|
The Structure, Function and Regulation of Protein Tyrosine Phosphatase Receptor Type J and Its Role in Diseases. Cells 2022; 12:cells12010008. [PMID: 36611803 PMCID: PMC9818648 DOI: 10.3390/cells12010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Protein tyrosine phosphatase receptor type J (PTPRJ), also known as DEP-1, HPTPη, or CD148, belongs to the R3 subfamily of receptor protein tyrosine phosphatases (RPTPs). It was first identified as an antioncogene due to its protein level being significantly downregulated in most epithelial tumors and cancer cell lines (e.g., colon, lung, thyroid, breast, and pancreas). PTPRJ regulates mouse optic nerve projection by inhibiting the phosphorylation of the erythropoietin-producing hepatocellular carcinoma (Eph) receptor and abelson murine leukemia viral oncogene homolog 1 (c-Abl). PTPRJ is crucial for metabolism. Recent studies have demonstrated that PTPRJ dephosphorylates JAK2 at positions Y813 and Y868 to inhibit leptin signaling. Akt is more phosphorylated at the Ser473 and Thr308 sites in Ptprj-/- mice, suggesting that PTPRJ may be a novel negative regulator of insulin signaling. PTPRJ also plays an important role in balancing the pro- and anti-osteoclastogenic activity of the M-CSF receptor (M-CSFR), and in maintaining NFATc1 expression during the late stages of osteoclastogenesis to promote bone-resorbing osteoclast (OCL) maturation. Furthermore, multiple receptor tyrosine kinases (RTKs) as substrates of PTPRJ are probably a potential therapeutic target for many types of diseases, such as cancer, neurodegenerative diseases, and metabolic diseases, by inhibiting their phosphorylation activity. In light of the important roles that PTPRJ plays in many diseases, this review summarizes the structural features of the protein, its expression pattern, and the physiological and pathological functions of PTPRJ, to provide new ideas for treating PTPRJ as a potential therapeutic target for related metabolic diseases and cancer.
Collapse
|
4
|
Wang Y, Zhou X, Lei Y, Chu Y, Yu X, Tong Q, Zhu T, Yu H, Fang S, Li G, Wang L, Wang GY, Xie X, Zhang J. NNMT contributes to high metastasis of triple negative breast cancer by enhancing PP2A/MEK/ERK/c-Jun/ABCA1 pathway mediated membrane fluidity. Cancer Lett 2022; 547:215884. [PMID: 35988817 DOI: 10.1016/j.canlet.2022.215884] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/12/2022] [Accepted: 08/12/2022] [Indexed: 11/02/2022]
Abstract
Elucidating the mechanism for high metastasis capacity of triple negative breast cancers (TNBC) is crucial to improve treatment outcomes of TNBC. We have recently reported that nicotinamide N-methyltransferase (NNMT) is overexpressed in breast cancer, especially in TNBC, and predicts poor survival of patients undergoing chemotherapy. Here, we aimed to determine the function and mechanism of NNMT on metastasis of TNBC. Additionally, analysis of public datasets indicated that NNMT is involved in cholesterol metabolism. In vitro, NNMT overexpression promoted migration and invasion of TNBCs by reducing cholesterol levels in the cytoplasm and cell membrane. Mechanistically, NNMT activated MEK/ERK/c-Jun/ABCA1 pathway by repressing protein phosphatase 2A (PP2A) activity leading to cholesterol efflux and membrane fluidity enhancement, thereby promoting the epithelial-mesenchymal transition (EMT) of TNBCs. In vivo, the metastasis capacity of TNBCs was weakened by targeting NNMT. Collectively, our findings suggest a new molecular mechanism involving NNMT in metastasis and poor survival of TNBC mediated by PP2A and affecting cholesterol metabolism.
Collapse
Affiliation(s)
- Yanzhong Wang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Clinical Laboratory, Xiasha Campus, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Xi Zhou
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Yinjiao Lei
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Yadong Chu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China; Department of Clinical Laboratory, Zhejiang Armed Police Corps Hospital, Hangzhou, Zhejiang, PR China
| | - Xingtong Yu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Qingchao Tong
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Tao Zhu
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Haitao Yu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Sining Fang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Guoli Li
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China
| | - Linbo Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Gavin Y Wang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, 29425, SC, USA; Cancer Cell Biology Program of the Hollings Cancer Center, Medical University of South Carolina, Charleston, 29425, SC, USA
| | - Xinyou Xie
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Clinical Laboratory, Xiasha Campus, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China.
| | - Jun Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Clinical Laboratory, Xiasha Campus, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
5
|
PTPRJ is downregulated in cervical squamous cell carcinoma. J Genet 2022. [DOI: 10.1007/s12041-022-01368-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
6
|
An HDAC9-associated immune-related signature predicts bladder cancer prognosis. PLoS One 2022; 17:e0264527. [PMID: 35239708 PMCID: PMC8893690 DOI: 10.1371/journal.pone.0264527] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/12/2022] [Indexed: 02/07/2023] Open
Abstract
Background The close relationship between histone deacetylase 9 (HDAC9) and immunity has attracted attention. We constructed an immune signature for HDAC9, a vital epigenetic modification, to predict the survival status and treatment benefits in bladder cancer (BC). Methods An exhaustive analysis of HDAC9 and immunology via the tumor and immune system interaction database (TISIDB) was performed, and an immune prognostic risk signature was developed based on genes enriched in the top five immune-related pathways under high HDAC9 status. Comprehensive analysis of survival curves and Cox regression were used to estimate the effectiveness of the risk signature. The relationship between immunological characteristics and the risk score was evaluated, and the mechanisms were also explored. Results In the TISIDB, HDAC9 was closely related to various immunological characteristics. The risk signature was obtained based on genes related to prognosis enriched in the top five immune-related pathways under high HDAC9 status. The survival rate of the high-risk BC patients was poor. The risk score was closely related to multiple immunological characteristics, drug sensitivity, immunotherapy benefits and biofunctions. Conclusion An immune-related prognostic signature established for HDAC9 expression status could independently predict the prognosis of BC patients. The use of this signature could help clinicians make personalized treatment decisions.
Collapse
|
7
|
Buenafe AC, Dorrell C, Reddy AP, Klimek J, Marks DL. Proteomic analysis distinguishes extracellular vesicles produced by cancerous versus healthy pancreatic organoids. Sci Rep 2022; 12:3556. [PMID: 35241737 PMCID: PMC8894448 DOI: 10.1038/s41598-022-07451-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/14/2022] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are produced and released by both healthy and malignant cells and bear markers indicative of ongoing biological processes. In the present study we utilized high resolution flow cytometry to detect EVs in the plasma of patients with pancreatic ductal adenocarcinoma (PDAC) and in the supernatants of PDAC and healthy control (HC) pancreatic organoid cultures. Using ultrafiltration and size exclusion chromatography, PDAC and HC pancreatic organoid EVs were isolated for mass spectrometry analysis. Proteomic and functional protein network analysis showed a striking distinction in that EV proteins profiled in pancreatic cancer organoids were involved in vesicular transport and tumorigenesis while EV proteins in healthy organoids were involved in cellular homeostasis. Thus, the most abundant proteins identified in either case represented non-overlapping cellular programs. Tumor-promoting candidates LAMA5, SDCBP and TENA were consistently upregulated in PDAC EVs. Validation of specific markers for PDAC EVs versus healthy pancreatic EVs will provide the biomarkers and enhanced sensitivity necessary to monitor early disease or disease progression, with or without treatment. Moreover, disease-associated changes in EV protein profiles provide an opportunity to investigate alterations in cellular programming with disease progression.
Collapse
Affiliation(s)
- Abigail C Buenafe
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR, USA.
| | - Craig Dorrell
- Oregon Stem Cell Center, Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR, USA
| | - Ashok P Reddy
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR, USA
| | - John Klimek
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
8
|
Protein Tyrosine Phosphatases: Mechanisms in Cancer. Int J Mol Sci 2021; 22:ijms222312865. [PMID: 34884670 PMCID: PMC8657787 DOI: 10.3390/ijms222312865] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Protein tyrosine kinases, especially receptor tyrosine kinases, have dominated the cancer therapeutics sphere as proteins that can be inhibited to selectively target cancer. However, protein tyrosine phosphatases (PTPs) are also an emerging target. Though historically known as negative regulators of the oncogenic tyrosine kinases, PTPs are now known to be both tumor-suppressive and oncogenic. This review will highlight key protein tyrosine phosphatases that have been thoroughly investigated in various cancers. Furthermore, the different mechanisms underlying pro-cancerous and anti-cancerous PTPs will also be explored.
Collapse
|
9
|
Chadar R, Kesharwani P. Nanotechnology-based siRNA delivery strategies for treatment of triple negative breast cancer. Int J Pharm 2021; 605:120835. [PMID: 34197908 DOI: 10.1016/j.ijpharm.2021.120835] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/13/2021] [Accepted: 06/25/2021] [Indexed: 12/26/2022]
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer characterized by absence of estrogen (ER) receptor, progesterone (PR) receptor, and human epidermal growth factor-2 (HER-2) receptor. TNBC is an aggressive disease that develops early Chemoresistance. The major pitfall associated is its poor prognosis, low overall survival, high relapse, and mortality as compared to other types of breast cancer. Chemotherapy could be helpful but do not contribute to an increase in survival of patient. To overcome such obstacles, in our article we explored advanced therapy using genes and nanocarrier along with its conjugation to achieve high therapeutic profile with reduced side effect. siRNAs are one of the class of RNA associated with gene silencing. They also regulate the expression of certain proteins that are involved in development of tumor cells. But they are highly unstable. So, for efficient delivery of siRNA, very intelligent, efficient delivery systems are required. Several nanotechnologies based non-viral vectors such as liposome, micelles, nanoparticles, dendrimers, exosomes, nanorods and nanobubbles etc. offers enormous unique properties such as nanometric size range, targeting potential with the capability to link with several targeting moieties for the gene delivery. These non-viral vectors are much safer, effective and efficient system for the delivery of genes along with chemotherapeutics. This review provides an overview of TNBC, conventional and advanced treatment approach of TNBC along with understanding of current status of several nanocarriers used for the delivery of siRNA for the treatment of TNBC.
Collapse
Affiliation(s)
- Rahul Chadar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
10
|
Young KA, Biggins L, Sharpe HJ. Protein tyrosine phosphatases in cell adhesion. Biochem J 2021; 478:1061-1083. [PMID: 33710332 PMCID: PMC7959691 DOI: 10.1042/bcj20200511] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023]
Abstract
Adhesive structures between cells and with the surrounding matrix are essential for the development of multicellular organisms. In addition to providing mechanical integrity, they are key signalling centres providing feedback on the extracellular environment to the cell interior, and vice versa. During development, mitosis and repair, cell adhesions must undergo extensive remodelling. Post-translational modifications of proteins within these complexes serve as switches for activity. Tyrosine phosphorylation is an important modification in cell adhesion that is dynamically regulated by the protein tyrosine phosphatases (PTPs) and protein tyrosine kinases. Several PTPs are implicated in the assembly and maintenance of cell adhesions, however, their signalling functions remain poorly defined. The PTPs can act by directly dephosphorylating adhesive complex components or function as scaffolds. In this review, we will focus on human PTPs and discuss their individual roles in major adhesion complexes, as well as Hippo signalling. We have collated PTP interactome and cell adhesome datasets, which reveal extensive connections between PTPs and cell adhesions that are relatively unexplored. Finally, we reflect on the dysregulation of PTPs and cell adhesions in disease.
Collapse
Affiliation(s)
- Katherine A. Young
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Laura Biggins
- Bioinformatics, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Hayley J. Sharpe
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, U.K
| |
Collapse
|
11
|
Sun Y, Li S, Yu W, Chen C, Liu T, Li L, Zhang D, Zhao Z, Gao J, Wang X, Shi D, Liu L. CD148 Serves as a Prognostic Marker of Gastric Cancer and Hinders Tumor Progression by Dephosphorylating EGFR. J Cancer 2020; 11:2667-2678. [PMID: 32201537 PMCID: PMC7065996 DOI: 10.7150/jca.40955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 01/24/2020] [Indexed: 01/06/2023] Open
Abstract
CD148 is a member of the receptor-type protein tyrosine phosphatase family encoded by the PTPRJ gene and has controversial impacts on cancers. In this study, we investigated the clinical significance of CD148 in gastric cancer and the possible mechanisms. Suppressed CD148 expression indicated adverse pathological features and poor outcomes in gastric cancer patients. CD148 overexpression impeded tumor proliferation, motility, and invasiveness, while CD148 knock-down or knockout promoted the ability of gastric cancer cells to grow and metastasize in vitro and in vivo. Mechanistically, CD148 negatively regulated EGFR phosphorylation of multiple tyrosine residues, including Y1173, Y1068, and Y1092, and remarkably inhibited downstream PI3K/AKT and MEK/ERK pathways. In silico analysis revealed that gene deletions or missense/truncated mutations of PTPRJ gene rarely occurred in gastric cancers. Instead, a 3' UTR-specific methylation might regulate CD148 expression, and the potential regulators were TET2 and TET3. Collectively, our results suggest that CD148 is a convincing prognostic marker as well as a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Yiting Sun
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.,Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Song Li
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Wenbin Yu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Cheng Chen
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Teng Liu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Lanbo Li
- Animal Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Di Zhang
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Zeyi Zhao
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Jing Gao
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiao Wang
- Department of Pathology, School of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Duanbo Shi
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Lian Liu
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
12
|
Ruckert MT, de Andrade PV, Santos VS, Silveira VS. Protein tyrosine phosphatases: promising targets in pancreatic ductal adenocarcinoma. Cell Mol Life Sci 2019; 76:2571-2592. [PMID: 30982078 PMCID: PMC11105579 DOI: 10.1007/s00018-019-03095-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 03/25/2019] [Accepted: 04/08/2019] [Indexed: 12/21/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer. It is the fourth leading cause of cancer-related death and is associated with a very poor prognosis. KRAS driver mutations occur in approximately 95% of PDAC cases and cause the activation of several signaling pathways such as mitogen-activated protein kinase (MAPK) pathways. Regulation of these signaling pathways is orchestrated by feedback loops mediated by the balance between protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs), leading to activation or inhibition of its downstream targets. The human PTPome comprises 125 members, and these proteins are classified into three distinct families according to their structure. Since PTP activity description, it has become clear that they have both inhibitory and stimulatory effects on cancer-associated signaling processes and that deregulation of PTP function is closely associated with tumorigenesis. Several PTPs have displayed either tumor suppressor or oncogenic characteristics during the development and progression of PDAC. In this sense, PTPs have been presented as promising candidates for the treatment of human pancreatic cancer, and many PTP inhibitors have been developed since these proteins were first associated with cancer. Nevertheless, some challenges persist regarding the development of effective and safe methods to target these molecules and deliver these drugs. In this review, we discuss the role of PTPs in tumorigenesis as tumor suppressor and oncogenic proteins. We have focused on the differential expression of these proteins in PDAC, as well as their clinical implications and possible targeting for pharmacological inhibition in cancer therapy.
Collapse
Affiliation(s)
- Mariana Tannús Ruckert
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil
| | - Pamela Viani de Andrade
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil
| | - Verena Silva Santos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil
| | - Vanessa Silva Silveira
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
13
|
Downregulation of PTPRK Promotes Cell Proliferation and Metastasis of NSCLC by Enhancing STAT3 Activation. Anal Cell Pathol (Amst) 2019; 2019:4265040. [PMID: 30838170 PMCID: PMC6374804 DOI: 10.1155/2019/4265040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/04/2018] [Accepted: 11/24/2018] [Indexed: 12/22/2022] Open
Abstract
Objective The receptor-type tyrosine-protein phosphatase κ (PTPRK) is a candidate tumor suppressor involved in the tumorigenesis of various organs. However, its expression and biological roles in non-small-cell lung cancer (NSCLC) have not yet been investigated. Methods PTPRK expression in NSCLC tissues and cell lines was examined using real-time PCR and western blotting. In addition, the effects of PTPRK on cell migration, invasion, and proliferation were evaluated in vitro. Furthermore, we explored whether the downregulation of PTPRK led to STAT3 activation in NSCLC cell lines by western blotting. The expression of phospho-STAT3Tyr705 in primary human NSCLC tissues was evaluated by immunohistochemistry. Results The results showed that PTPRK expression was frequently reduced in NSCLC tissues with lymph node metastasis and cell lines. The inhibition of PTPRK expression resulted in increased proliferation, invasion, and migration of NSCLC cells in vitro. Additionally, after silencing of PTPRK, phospho-STAT3Tyr705 was significantly increased in NSCLC cells. Moreover, the phospho-STAT3Tyr705 levels of NSCLC tissues were positively correlated with lymph node metastasis and significantly inversely correlated with the expression of PTPRK (p < 0.05). Conclusions These results suggested that PTPRK functions as a novel tumor suppressor in NSCLC, and its suppressive ability may be involved in STAT3 activation.
Collapse
|
14
|
Meta-analysis of association between Arg326Gln (rs1503185) and Gln276Pro (rs1566734) polymorphisms of PTPRJ gene and cancer risk. J Appl Genet 2019; 60:57-62. [PMID: 30661225 PMCID: PMC6373398 DOI: 10.1007/s13353-019-00481-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/21/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022]
Abstract
Protein tyrosine phosphatase receptor type J (PTPRJ, DEP1) is a tumour suppressor gene that negatively regulates such processes as angiogenesis, cell proliferation and migration and is one of the genes important for tumour development. Similar to other phosphatase genes, PTPRJ is also described as an oncogene. Among various genetic changes characteristic for this gene, single nucleotide polymorphisms (SNPs) constituting benign genetic variants that can modulate its function have been described. We focused on Gln276Pro and Arg326Gln missense polymorphisms and performed a meta-analysis using data from 2930 and 852 patients for Gln276Pro and Arg326Gln respectively in different cancers. A meta-analysis was performed based on five articles accessed via the PubMed and Research Gate databases. Our meta-analysis revealed that for Arg326Gln, the presence of the Arg (C) allele was associated with lower risk of some cancers, the strongest association was observed for colorectal cancer patients, and there was no association between Gln276Pro (G>T) polymorphism and cancer risk. The polymorphisms Arg326Gln and Gln276Pro of the PTPRJ gene are not associated with an increased risk of cancer except for the Arg326Gln polymorphism in colorectal cancer. Large-scale studies should be performed to verify the impact of this SNP on individual susceptibility to colorectal cancer for given individuals.
Collapse
|
15
|
Wang YH, Yin YW, Zhou H, Cao YD. miR-639 is associated with advanced cancer stages and promotes proliferation and migration of nasopharyngeal carcinoma. Oncol Lett 2018; 16:6903-6909. [PMID: 30546422 PMCID: PMC6256336 DOI: 10.3892/ol.2018.9512] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/31/2017] [Indexed: 01/29/2023] Open
Abstract
Early detection of nasopharyngeal carcinoma (NPC) is of vital importance for improving prognosis and survival rates. MicroRNA (miRNA) are a class of short and non-coding RNA molecules that are capable of inhibiting the translation of mRNA of target genes. Previous studies have revealed that miRNA are involved in tumorigenesis and cancer development. The RNase-resistance of circulating miRNA have made them valuable non-invasive biomarkers, and has therefore drawn particular attention to their therapeutic potential. The aim of the present study was to investigate the expression of the previously uncharacterized miR-639 in NPC. In a study population of 139 patients, higher expression of miR-639 was associated with metastasis, more advanced cancer stages, and lower disease-free survival rates. In vitro experiments involving transfection of human NPC C666-1 and NPC/HK1 cell lines with miR-639 mimics and antagomir indicated that overexpressing miR-639 promoted cell proliferation and migration, suppression of miR-639 inhibited proliferation and migration. The present study provides evidence that miR-639 is differentially expressed in NPC tissues of varying cancer stages, and suggests that quantifying circulating miR-639 may be of importance for non-invasive diagnosis and prognostic evaluation, and may have potential therapeutic utility.
Collapse
Affiliation(s)
- Yun-Hui Wang
- Department of Ear, Nose and Throat, Linyi City People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Yan-Wei Yin
- Department of Oncology, Linyi City People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Han Zhou
- Department of Ear, Nose and Throat, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yuan-Dong Cao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
16
|
Hendricks WPD, Zismann V, Sivaprakasam K, Legendre C, Poorman K, Tembe W, Perdigones N, Kiefer J, Liang W, DeLuca V, Stark M, Ruhe A, Froman R, Duesbery NS, Washington M, Aldrich J, Neff MW, Huentelman MJ, Hayward N, Brown K, Thamm D, Post G, Khanna C, Davis B, Breen M, Sekulic A, Trent JM. Somatic inactivating PTPRJ mutations and dysregulated pathways identified in canine malignant melanoma by integrated comparative genomic analysis. PLoS Genet 2018; 14:e1007589. [PMID: 30188888 PMCID: PMC6126841 DOI: 10.1371/journal.pgen.1007589] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/24/2018] [Indexed: 01/11/2023] Open
Abstract
Canine malignant melanoma, a significant cause of mortality in domestic dogs, is a powerful comparative model for human melanoma, but little is known about its genetic etiology. We mapped the genomic landscape of canine melanoma through multi-platform analysis of 37 tumors (31 mucosal, 3 acral, 2 cutaneous, and 1 uveal) and 17 matching constitutional samples including long- and short-insert whole genome sequencing, RNA sequencing, array comparative genomic hybridization, single nucleotide polymorphism array, and targeted Sanger sequencing analyses. We identified novel predominantly truncating mutations in the putative tumor suppressor gene PTPRJ in 19% of cases. No BRAF mutations were detected, but activating RAS mutations (24% of cases) occurred in conserved hotspots in all cutaneous and acral and 13% of mucosal subtypes. MDM2 amplifications (24%) and TP53 mutations (19%) were mutually exclusive. Additional low-frequency recurrent alterations were observed amidst low point mutation rates, an absence of ultraviolet light mutational signatures, and an abundance of copy number and structural alterations. Mutations that modulate cell proliferation and cell cycle control were common and highlight therapeutic axes such as MEK and MDM2 inhibition. This mutational landscape resembles that seen in BRAF wild-type and sun-shielded human melanoma subtypes. Overall, these data inform biological comparisons between canine and human melanoma while suggesting actionable targets in both species.
Collapse
Affiliation(s)
- William P. D. Hendricks
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Victoria Zismann
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Karthigayini Sivaprakasam
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Department of Biomedical Informatics, Arizona State University, Phoenix, Arizona, United States of America
| | - Christophe Legendre
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Kelsey Poorman
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States of America
- Department of Dermatology, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Waibhav Tembe
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Nieves Perdigones
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Jeffrey Kiefer
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Winnie Liang
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Valerie DeLuca
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- School of Life Sciences, Arizona State University, Phoenix, Arizona, United States of America
| | - Mitchell Stark
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Alison Ruhe
- Veterinary Genetics Laboratory, University of California Davis, Davis, California, United States of America
| | - Roe Froman
- Laboratory of Cancer and Developmental Cell Biology, Van Andel Research Institute (VARI), Grand Rapids, Michigan, United States of America
| | | | - Megan Washington
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Jessica Aldrich
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Mark W. Neff
- Program in Canine Genetics and Genomics, Van Andel Research Institute (VARI), Grand Rapids, Michigan, United States of America
| | - Matthew J. Huentelman
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Nicholas Hayward
- Oncogenomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Kevin Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Douglas Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, United States of America
| | - Gerald Post
- The Veterinary Cancer Center, Norwalk, Connecticut, United States of America
| | - Chand Khanna
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Barbara Davis
- Innogenics Inc., Harvard, Massachusetts, United States of America
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States of America
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States of America
| | - Alexander Sekulic
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Department of Dermatology, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Jeffrey M. Trent
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| |
Collapse
|
17
|
Narla G, Sangodkar J, Ryder CB. The impact of phosphatases on proliferative and survival signaling in cancer. Cell Mol Life Sci 2018; 75:2695-2718. [PMID: 29725697 PMCID: PMC6023766 DOI: 10.1007/s00018-018-2826-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/24/2018] [Accepted: 04/23/2018] [Indexed: 02/06/2023]
Abstract
The dynamic and stringent coordination of kinase and phosphatase activity controls a myriad of physiologic processes. Aberrations that disrupt the balance of this interplay represent the basis of numerous diseases. For a variety of reasons, early work in this area portrayed kinases as the dominant actors in these signaling events with phosphatases playing a secondary role. In oncology, these efforts led to breakthroughs that have dramatically altered the course of certain diseases and directed vast resources toward the development of additional kinase-targeted therapies. Yet, more recent scientific efforts have demonstrated a prominent and sometimes driving role for phosphatases across numerous malignancies. This maturation of the phosphatase field has brought with it the promise of further therapeutic advances in the field of oncology. In this review, we discuss the role of phosphatases in the regulation of cellular proliferation and survival signaling using the examples of the MAPK and PI3K/AKT pathways, c-Myc and the apoptosis machinery. Emphasis is placed on instances where these signaling networks are perturbed by dysregulation of specific phosphatases to favor growth and persistence of human cancer.
Collapse
Affiliation(s)
| | - Jaya Sangodkar
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
18
|
Combinational siRNA delivery using hyaluronic acid modified amphiphilic polyplexes against cell cycle and phosphatase proteins to inhibit growth and migration of triple-negative breast cancer cells. Acta Biomater 2018; 66:294-309. [PMID: 29183848 DOI: 10.1016/j.actbio.2017.11.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/05/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022]
Abstract
Triple-negative breast cancer is an aggressive form of breast cancer with few therapeutic options if it recurs after adjuvant chemotherapy. RNA interference could be an alternative therapy for metastatic breast cancer, where small interfering RNA (siRNA) can silence the expression of aberrant genes critical for growth and migration of malignant cells. Here, we formulated a siRNA delivery system using lipid-substituted polyethylenimine (PEI) and hyaluronic acid (HA), and characterized the size, ζ-potential and cellular uptake of the nanoparticulate delivery system. Higher cellular uptake of siRNA by the tailored PEI/HA formulation suggested better interaction of complexes with breast cancer cells due to improved physicochemical characteristics of carrier and HA-binding CD44 receptors. The siRNAs against specific phosphatases that inhibited migration of MDA-MB-231 cells were then identified using library screen against 267 protein-tyrosine phosphatases, and siRNAs to inhibit cell migration were further validated. We then assessed the combinational delivery of a siRNA against CDC20 to decrease cell growth and a siRNA against several phosphatases shown to decrease migration of breast cancer cells. Combinational siRNA therapy against CDC20 and identified phosphatases PPP1R7, PTPN1, PTPN22, LHPP, PPP1R12A and DUPD1 successfully inhibited cell growth and migration, respectively, without interfering the functional effect of the co-delivered siRNA. The identified phosphatases could serve as potential targets to inhibit migration of highly aggressive metastatic breast cancer cells. Combinational siRNA delivery against cell cycle and phosphatases could be a promising strategy to inhibit both growth and migration of metastatic breast cancer cells, and potentially other types of metastatic cancer. STATEMENT OF SIGNIFICANCE The manuscript investigated the efficacy of a tailored polymeric siRNA delivery system formulation as well as combinational siRNA therapy in metastatic breast cancer cells to inhibit malignant cell growth and migration. The siRNA delivery was undertaken by non-viral means with PEI/HA. We identified six phosphatases that could be critical targets to inhibit migration of highly aggressive metastatic breast cancer cells. We further report on specifically targeting cell cycle and phosphatase proteins to decrease both malignant cell growth and migration simultaneously. Clinical gene therapy against metastatic breast cancer with effective and safe delivery systems is urgently needed to realize the potential of molecular medicine in this deadly disease and our studies in this manuscript is intended to facilitate this endeavor.
Collapse
|
19
|
Meeusen B, Janssens V. Tumor suppressive protein phosphatases in human cancer: Emerging targets for therapeutic intervention and tumor stratification. Int J Biochem Cell Biol 2017; 96:98-134. [PMID: 29031806 DOI: 10.1016/j.biocel.2017.10.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
Aberrant protein phosphorylation is one of the hallmarks of cancer cells, and in many cases a prerequisite to sustain tumor development and progression. Like protein kinases, protein phosphatases are key regulators of cell signaling. However, their contribution to aberrant signaling in cancer cells is overall less well appreciated, and therefore, their clinical potential remains largely unexploited. In this review, we provide an overview of tumor suppressive protein phosphatases in human cancer. Along their mechanisms of inactivation in defined cancer contexts, we give an overview of their functional roles in diverse signaling pathways that contribute to their tumor suppressive abilities. Finally, we discuss their emerging roles as predictive or prognostic markers, their potential as synthetic lethality targets, and the current feasibility of their reactivation with pharmacologic compounds as promising new cancer therapies. We conclude that their inclusion in clinical practice has obvious potential to significantly improve therapeutic outcome in various ways, and should now definitely be pushed forward.
Collapse
Affiliation(s)
- Bob Meeusen
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium.
| |
Collapse
|
20
|
Elson A. Stepping out of the shadows: Oncogenic and tumor-promoting protein tyrosine phosphatases. Int J Biochem Cell Biol 2017; 96:135-147. [PMID: 28941747 DOI: 10.1016/j.biocel.2017.09.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 12/18/2022]
Abstract
Protein tyrosine phosphorylation is critical for proper function of cells and organisms. Phosphorylation is regulated by the concerted but generically opposing activities of tyrosine kinases (PTKs) and tyrosine phosphatases (PTPs), which ensure its proper regulation, reversibility, and ability to respond to changing physiological situations. Historically, PTKs have been associated mainly with oncogenic and pro-tumorigenic activities, leading to the generalization that protein dephosphorylation is anti-oncogenic and hence that PTPs are tumor-suppressors. In many cases PTPs do suppress tumorigenesis. However, a growing body of evidence indicates that PTPs act as dominant oncogenes and drive cell transformation in a number of contexts, while in others PTPs support transformation that is driven by other oncogenes. This review summarizes the known transforming and tumor-promoting activities of the classical, tyrosine specific PTPs and highlights their potential as drug targets for cancer therapy.
Collapse
Affiliation(s)
- Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
21
|
Zhang XF, Tu R, Li K, Ye P, Cui X. Tumor Suppressor PTPRJ Is a Target of miR-155 in Colorectal Cancer. J Cell Biochem 2017; 118:3391-3400. [PMID: 28316102 DOI: 10.1002/jcb.25995] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/16/2017] [Indexed: 01/02/2023]
Abstract
PTPRJ is known for its antiproliferative role. Loss of heterozygosity (LOH) of PTPRJ has frequently been observed in various human cancers including colorectal cancer (CRC), lung cancer, and breast cancer. However, the function and mechanism of PTPRJ in CRC are not well understood. At the present study, we show that ectopic expression of PTPRJ inhibits cell growth, migration, and invasiveness in CRC cell line HCT116. Moreover, PTPRJ inhibits the tumorigenecity of HCT116 in a xenograft tumor model. MiR-155, the well-known oncomiR in CRC, is identified as an upstream factor of PTPRJ. MiR-155 directly binds to the 3' untranslated region of PTPRJ mRNA and suppresses the mRNA and protein levels of PTPRJ. Furthermore, the growth-promoting and AKT signaling activation effect of miR-155 was abrogated by PTPRJ overexpression, and vice versa. Our study reveals the crucial role of miR-155/PTPRJ/AKT axis in proliferation and migration of CRC cells and suggests a therapeutic potential of PTPRJ. J. Cell. Biochem. 118: 3391-3400, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiao-Fei Zhang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430072, P. R. China
| | - Rongfu Tu
- College of Life Sciences, Wuhan University, Wuhan 430070, P. R. China
| | - Keke Li
- College of Life Sciences, Wuhan University, Wuhan 430070, P. R. China
| | - Pengxiang Ye
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430072, P. R. China
| | - Xiaofeng Cui
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430072, P. R. China
| |
Collapse
|
22
|
Sun PH, Chen G, Mason M, Jiang WG, Ye L. Dual roles of protein tyrosine phosphatase kappa in coordinating angiogenesis induced by pro-angiogenic factors. Int J Oncol 2017; 50:1127-1135. [PMID: 28259897 PMCID: PMC5363875 DOI: 10.3892/ijo.2017.3884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 01/18/2017] [Indexed: 11/09/2022] Open
Abstract
A potential role may be played by receptor-type protein tyrosine phosphatase kappa (PTPRK) in angiogenesis due to its critical function in coordinating intracellular signal transduction from various receptors reliant on tyrosine phosphorylation. In the present study, we investigated the involvement of PTPRK in the cellular functions of vascular endothelial cells (HECV) and its role in angiogenesis using in vitro assays and a PTPRK knockdown vascular endothelial cell model. PTPRK knockdown in HECV cells (HECVPTPRKkd) resulted in a decrease of cell proliferation and cell-matrix adhesion; however, increased cell spreading and motility were seen. Reduced focal adhesion kinase (FAK) and paxillin protein levels were seen in the PTPRK knockdown cells which may contribute to the inhibitory effect on adhesion. HECVPTPRKkd cells were more responsive to the treatment of fibroblast growth factor (FGF) in their migration compared with the untreated control and cells treated with VEGF. Moreover, elevated c-Src and Akt1 were seen in the PTPRK knockdown cells. The FGF-promoted cell migration was remarkably suppressed by an addition of PLCγ inhibitor compared with other small inhibitors. Knockdown of PTPRK suppressed the ability of HECV cells to form tubules and also impaired the tubule formation that was induced by FGF and conditioned medium of cancer cells. Taken together, it suggests that PTPRK plays dual roles in coordinating angiogenesis. It plays a positive role in cell proliferation, adhesion and tubule formation, but suppresses cell migration, in particular, the FGF-promoted migration. PTPRK bears potential to be targeted for the prevention of tumour associated angiogenesis.
Collapse
Affiliation(s)
- Ping-Hui Sun
- Cardiff China Medical Research Collaborative Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Gang Chen
- Cardiff China Medical Research Collaborative Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Malcolm Mason
- Cardiff China Medical Research Collaborative Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| | - Lin Ye
- Cardiff China Medical Research Collaborative Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, CF14 4XN, UK
| |
Collapse
|
23
|
Watkins RJ, Imruetaicharoenchoke W, Read ML, Sharma N, Poole VL, Gentilin E, Bansal S, Bosseboeuf E, Fletcher R, Nieto HR, Mallick U, Hackshaw A, Mehanna H, Boelaert K, Smith VE, McCabe CJ. Pro-invasive Effect of Proto-oncogene PBF Is Modulated by an Interaction with Cortactin. J Clin Endocrinol Metab 2016; 101:4551-4563. [PMID: 27603901 PMCID: PMC5155689 DOI: 10.1210/jc.2016-1932] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Metastatic disease is responsible for the majority of endocrine cancer deaths. New therapeutic targets are urgently needed to improve patient survival rates. OBJECTIVE The proto-oncogene PTTG1-binding factor (PBF/PTTG1IP) is overexpressed in multiple endocrine cancers and circumstantially associated with tumor aggressiveness. This study aimed to understand the role of PBF in tumor cell invasion and identify possible routes to inhibit its action. Design, Setting, Patients, and Interventions: Thyroid, breast, and colorectal cells were transfected with PBF and cultured for in vitro analysis. PBF and cortactin (CTTN) expression was determined in differentiated thyroid cancer and The Cancer Genome Atlas RNA-seq data. PRIMARY OUTCOME MEASURE Pro-invasive effects of PBF were evaluated by 2D Boyden chamber, 3D organotypic, and proximity ligation assays. RESULTS Our study identified that PBF and CTTN physically interact and co-localize, and that this occurs at the cell periphery, particularly at the leading edge of migrating cancer cells. Critically, PBF induces potent cellular invasion and migration in thyroid and breast cancer cells, which is entirely abrogated in the absence of CTTN. Importantly, we found that CTTN is over-expressed in differentiated thyroid cancer, particularly in patients with regional lymph node metastasis, which significantly correlates with elevated PBF expression. Mutation of PBF (Y174A) or pharmacological intervention modulates the PBF: CTTN interaction and attenuates the invasive properties of cancer cells. CONCLUSION Our results demonstrate a unique role for PBF in regulating CTTN function to promote endocrine cell invasion and migration, as well as identify a new targetable interaction to block tumor cell movement.
Collapse
Affiliation(s)
- Rachel J Watkins
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Waraporn Imruetaicharoenchoke
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Martin L Read
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Neil Sharma
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Vikki L Poole
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Erica Gentilin
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Sukhchain Bansal
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Emy Bosseboeuf
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Rachel Fletcher
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Hannah R Nieto
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Ujjal Mallick
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Allan Hackshaw
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Hisham Mehanna
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Kristien Boelaert
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Vicki E Smith
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Christopher J McCabe
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
24
|
Qi Y, Dai Y, Gui S. Protein tyrosine phosphatase PTPRB regulates Src phosphorylation and tumour progression in NSCLC. Clin Exp Pharmacol Physiol 2016; 43:1004-12. [PMID: 27314562 DOI: 10.1111/1440-1681.12610] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Yinliang Qi
- Department of Respiratory Disease; Anhui Medical University; Hefei Anhui China
- General Department of Hyperbaric Oxygen; The Second People's Hospital of Hefei; Hefei Anhui China
| | - Yuanchang Dai
- General Department of Hyperbaric Oxygen; The Second People's Hospital of Hefei; Hefei Anhui China
| | - Shuyu Gui
- Department of Respiratory Disease; Anhui Medical University; Hefei Anhui China
| |
Collapse
|
25
|
Fournier P, Dussault S, Fusco A, Rivard A, Royal I. Tyrosine Phosphatase PTPRJ/DEP-1 Is an Essential Promoter of Vascular Permeability, Angiogenesis, and Tumor Progression. Cancer Res 2016; 76:5080-91. [PMID: 27364551 DOI: 10.1158/0008-5472.can-16-1071] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/24/2016] [Indexed: 11/16/2022]
Abstract
The protein tyrosine phosphatase PTPRJ/DEP-1 has been implicated in negative growth regulation in endothelial cells, where its expression varies at transitions between proliferation and contact inhibition. However, in the same cells, DEP-1 has also been implicated in VEGF-dependent Src activation, permeability, and capillary formation, suggesting a positive role in regulating these functions. To resolve this dichotomy in vivo, we investigated postnatal angiogenesis and vascular permeability in a DEP-1-deficient mouse. In this study, we report that DEP-1 is required for Src activation and phosphorylation of its endothelial cell-specific substrate, VE-cadherin, after systemic injection of VEGF. Accordingly, VEGF-induced vascular leakage was abrogated in the DEP-1-deficient mice. Furthermore, capillary formation was impaired in murine aortic tissue rings or Matrigel plugs infused with VEGF. In the absence of DEP-1, angiogenesis triggered by ischemia or during tumor formation was defective, which in the latter case was associated with reduced tumor cell proliferation and increased apoptosis. Macrophage infiltration was also impaired, reflecting reduced vascular permeability in the tumors or a possible cell autonomous effect of DEP-1. Consequently, the formation of spontaneous and experimental lung metastases was strongly decreased in DEP-1-deficient mice. In clinical specimens of cancer, less vascularized tumors exhibited lower microvascular expression of DEP-1. Altogether, our results established DEP-1 as an essential driver of VEGF-dependent permeability, angiogenesis, and metastasis, suggesting a novel therapeutic route to cancer treatment. Cancer Res; 76(17); 5080-91. ©2016 AACR.
Collapse
Affiliation(s)
- Patrick Fournier
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada. Institut du cancer de Montréal, Montréal, Quebec, Canada
| | - Sylvie Dussault
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
| | - Alfredo Fusco
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Alain Rivard
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada. Département de Médecine, Université de Montréal, Montréal, Quebec, Canada
| | - Isabelle Royal
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada. Institut du cancer de Montréal, Montréal, Quebec, Canada. Département de Médecine, Université de Montréal, Montréal, Quebec, Canada.
| |
Collapse
|
26
|
Zhang M, Song S, Yi Z, Zhao X, Fu L, Wang L, Ma C, Mao M, Xing Y, Zhu D. Human biliverdin reductase promotes EMT through the ERK1/2 signal pathway in breast cancer. Eur J Pharmacol 2016; 788:45-53. [PMID: 27316791 DOI: 10.1016/j.ejphar.2016.06.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 06/04/2016] [Accepted: 06/13/2016] [Indexed: 12/20/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) plays an important role in the development of the invasive and metastatic potentials of breast cancer cells during progression. Human biliverdin reductase (hBVR), an enzyme in the heme metabolism pathway, is involved in hypoxia-induced renal tubular EMT. However, whether hBVR contributes to the EMT of breast cancer remains unclear. Here, we used breast cancer cell lines (MCF-7, T-47D) and normal breast epithelial cells (MCF-10A) to explore the potential role of hBVR in the EMT of breast cancer. Western blot, RT-PCR and immunofluorescence were employed to test the expression and location of hBVR in the cell lines. Small interfering RNA of hBVR (si-hBVR) was used to knockdown the expression of hBVR, and U0126 was applied to inhibit the ERK1/2 signaling in MCF-7, T-47D cells. We found that hBVR highly expressed in MCF-7 and T-47D cells compared with MCF-10A cells, and had different cellular locations between them. Our results revealed that EMT occurred in tissues from breast cancer patients and breast cancer cell lines. However, the EMT in MCF-7 and T-47D cells was suppressed by si-hBVR and U0126. Furthermore, the expression of phosphorylated ERK1/2 was down-regulated by si-hBVR. In addition, hBVR regulated EMT through the ERK1/2 signaling, but bilirubin, which is a product of hBVR in the heme metabolism pathway in breast cancer, did not. Taken together, these findings provide new evidence that hBVR plays an important role in promoting EMT in human breast cancer through the ERK1/2 signaling pathway, and hBVR may be a therapeutic target for this disease.
Collapse
Affiliation(s)
- Min Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Daqing 163319, China
| | - Shasha Song
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Daqing 163319, China
| | - Zhi Yi
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Daqing 163319, China
| | - Xijuan Zhao
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Daqing 163319, China
| | - Li Fu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Daqing 163319, China
| | - Lin Wang
- Department of Pathology, Harbin Medical University, No. 39 Xinyang, Daqing 163319, China
| | - Cui Ma
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Daqing 163319, China; Department of Immunology, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing 163319, China
| | - Min Mao
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Daqing 163319, China
| | - Yan Xing
- Department of Pharmacology, College of Basic Medicine, Harbin Medical University, Daqing 163319, China
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Daqing 163319, China; Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin 150081, China.
| |
Collapse
|
27
|
Loss of Protein Tyrosine Phosphatase Receptor J Expression Predicts an Aggressive Clinical Course in Patients with Esophageal Squamous Cell Carcinoma. Pathol Oncol Res 2015; 22:541-7. [DOI: 10.1007/s12253-015-0036-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/15/2015] [Indexed: 01/01/2023]
|
28
|
Luo X, Yang S, Zhou C, Pan F, Li Q, Ma S. MicroRNA-328 enhances cellular motility through posttranscriptional regulation of PTPRJ in human hepatocellular carcinoma. Onco Targets Ther 2015; 8:3159-67. [PMID: 26604785 PMCID: PMC4630182 DOI: 10.2147/ott.s93056] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Interaction between microRNA (miR-328) and PTPRJ (protein tyrosine phosphatase, receptor type, J) has been reported to be responsible for miR-328-dependent increase in epithelial cancer cell proliferation. However, the role of miR-328 and PTPRJ in hepatocellular carcinoma (HCC) remains unclear. The aim of this study was to investigate the clinical significance of miR-328 and/or PTPRJ expression in human HCC and determine their precise biological functions in this malignancy. METHODS Expression levels of miR-328 and PTPRJ messenger RNA (mRNA) in 100 pairs of HCC and adjacent noncancerous tissues were detected using quantitative real-time reverse transcription polymerase chain reaction. The associations between miR-328 and/or PTPRJ expression and various clinicopathological features of HCC patients were further statistically assessed. Then, the functions of miR-328 and PTPRJ in migration and invasion of two human HCC cell lines were determined by transwell assays. RESULTS miR-328 and PTPRJ mRNA expression levels were markedly upregulated and down-regulated in HCC tissues, respectively, compared to adjacent noncancerous tissues. Notably, the upregulation of miR-328 in HCC tissues was significantly correlated with the downregulation of PTPRJ mRNA in HCC tissues (r=-0.362, P=0.01). In addition, miR-328-high and/or PTPRJ-low expression were found to be closely correlated with high Edmondson-Steiner grading (all P<0.05) and advanced tumor-node-metastasis stage (all P<0.05). Moreover, the restoration of miR-328 dramatically promoted HCC cell migration and invasion by repressing PTPRJ expression. Interestingly, the loss of PTPRJ expression could significantly attenuate the inhibitory effects of knockdown miR-328 on the migration and invasion of HCC cells. CONCLUSION These findings demonstrated that the dysregulation of miR-328 and PTPRJ may be associated with tumor progression of HCC patients. Functionally, miR-328 may serve as a crucial oncogene and be implicated in the motility of HCC cells at least in part by the suppression of PTPRJ.
Collapse
Affiliation(s)
- Xiaoling Luo
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, People's Republic of China
| | - Shiyan Yang
- Department of Gastroenterology, Huai'an Hospital Affiliated to Xuzhou Medical College and Huai'an Second People's Hospital, Huai'an, Jiangsu, People's Republic of China
| | - Chuanwen Zhou
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, People's Republic of China
| | - Feng Pan
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, People's Republic of China
| | - Qianjun Li
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, People's Republic of China
| | - Shijie Ma
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, People's Republic of China
| |
Collapse
|