1
|
Yang Y, Li XM, Wang JR, Li Y, Ye WL, Wang Y, Liu YX, Deng ZY, Gan WJ, Wu H. TRIP6 promotes inflammatory damage via the activation of TRAF6 signaling in a murine model of DSS-induced colitis. J Inflamm (Lond) 2022; 19:1. [PMID: 34983535 PMCID: PMC8725398 DOI: 10.1186/s12950-021-00298-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/25/2021] [Indexed: 11/20/2022] Open
Abstract
Background TRIP6 is a zyxin family member that serves as an adaptor protein to regulate diverse biological processes. In prior reports, TRIP6 was shown to play a role in regulating inflammation. However, its in vivo roles and mechanistic importance in colitis remain largely elusive. Herein, we therefore employed TRIP6-deficient (TRIP6−/−) mice in order to explore the mechanistic importance of TRIP6 in a dextran sodium sulfate (DSS)-induced model of murine colitis. Findings Wild-type (TRIP6+/+) mice developed more severe colitis following DSS-mediated disease induction relative to TRIP6−/− mice, as evidenced by more severe colonic inflammation and associated crypt damage. TRIP6 expression in wild-type mice was significantly elevated following DSS treatment. Mechanistically, TRIP6 binds to TRAF6 and enhances oligomerization and autoubiquitination of TRAF6. This leads to the activation of NF-κB signaling and the expression of pro-inflammatory cytokines such as TNFα and IL-6, in the in vivo mouse model of colitis. Conclusions These in vivo data demonstrate that TRIP6 serves as a positive regulator of DSS-induced colitis through interactions with TRAF6 resulting in the activation of inflammatory TRAF6 signaling, highlighting its therapeutic promise as a protein that theoretically can be targeted to prevent or treat colitis. Supplementary Information The online version contains supplementary material available at 10.1186/s12950-021-00298-0.
Collapse
Affiliation(s)
- Yun Yang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou, 215123, China
| | - Xiu-Ming Li
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jing-Ru Wang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yan Li
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou, 215123, China
| | - Wen-Long Ye
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yi Wang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yu-Xuan Liu
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou, 215123, China
| | - Zhi-Yong Deng
- Department of Pathology, The First People's Hospital of Kunshan, Kunshan, Suzhou, 215300, China.
| | - Wen-Juan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated of Soochow University, Suzhou, 215124, China.
| | - Hua Wu
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou, 215123, China. .,Department of Pathology, Dushu Lake Hospital Affiliated of Soochow University, Suzhou, 215124, China.
| |
Collapse
|
2
|
Fatemipour M, Nahand JS, Fard Azar ME, Baghi HB, Taghizadieh M, Sorayyayi S, Hussen BM, Mirzaei H, Moghoofei M, Bokharaei-Salim F. Human papillomavirus and prostate cancer: The role of viral expressed proteins in the inhibition of anoikis and induction of metastasis. Microb Pathog 2021; 152:104576. [PMID: 33086103 DOI: 10.1016/j.micpath.2020.104576] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The aim of this study is to address the role of HPV in prostate cancer (PCa) development through the inducement of resistance to anoikis. METHODS In this case-control study, prostate tissues and blood samples were collected from 116 individuals, including 72 cases with PCa and 44 non-malignant prostate tissue samples as a control group. The expression level of HPV genes (E2, E6, and E7) and cellular genes including anti-apoptotic mediators (Bcl-2 and survivin), tumor suppressor proteins (Rb and p53), and some mediators involved in anoikis resistance and invasiveness (E-cadherin, N-cadherin, Twist, PTPN13 and SLUG) were evaluated. RESULTS HPV genome was identified in 36.1% cases and 15.9% control samples, additionally there was found to be a statistic significant association between the presence of HPV and PCa (OR = 1.64, 95% C.I = 0.8-1.8, P-value = 0.023). HPV genotype 16 and 18 were the most prevalent genotype in both in the PCa group and the control group. The expression level of the tumor suppressor proteins (Rb and p53) and anti-apoptotic mediators (Bcl-2 and Survivin) were significantly decreased and increased, respectively, in the HPV-positive specimens compared to the HPV-negative specimens. Furthermore, the mean expression level of N-cadherin, SLUG, and TWIST in the HPV-positive specimens was higher than HPV-negative specimens while the mean expression level of PTPN-13 and E-cadherin genes in the HPV-positive specimens was lower than HPV-negative specimens. CONCLUSION Our study suggests that HPV infection may be involved in the development of PCa metastases by modulating anoikis resistance related genes.
Collapse
Affiliation(s)
- Maryam Fatemipour
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, School of Medicine, Center for Women's Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Sorayyayi
- Department of Clinical Biochemistry, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Farah Bokharaei-Salim
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Dual Role of the PTPN13 Tyrosine Phosphatase in Cancer. Biomolecules 2020; 10:biom10121659. [PMID: 33322542 PMCID: PMC7763032 DOI: 10.3390/biom10121659] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 02/08/2023] Open
Abstract
In this review article, we present the current knowledge on PTPN13, a class I non-receptor protein tyrosine phosphatase identified in 1994. We focus particularly on its role in cancer, where PTPN13 acts as an oncogenic protein and also a tumor suppressor. To try to understand these apparent contradictory functions, we discuss PTPN13 implication in the FAS and oncogenic tyrosine kinase signaling pathways and in the associated biological activities, as well as its post-transcriptional and epigenetic regulation. Then, we describe PTPN13 clinical significance as a prognostic marker in different cancer types and its impact on anti-cancer treatment sensitivity. Finally, we present future research axes following recent findings on its role in cell junction regulation that implicate PTPN13 in cell death and cell migration, two major hallmarks of tumor formation and progression.
Collapse
|
4
|
Wu XN, Ma YY, Hao ZC, Wang H. [Research progress on the biological regulatory function of lysophosphatidic acid in bone tissue cells]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2020; 38:324-329. [PMID: 32573143 DOI: 10.7518/hxkq.2020.03.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Lysophosphatidic acid (LPA) is a small phospholipid that is present in all eukaryotic tissues and blood plasma. As an extracellular signaling molecule, LPA mediates many cellular functions by binding to six known G protein-coupled receptors and activating their downstream signaling pathways. These functions indicate that LPA may play important roles in many biological processes that include organismal development, wound healing, and carcinogenesis. Recently, many studies have found that LPA has various biological effects in different kinds of bone cells. These findings suggest that LPA is a potent regulator of bone development and remodeling and holds promising application potential in bone tissue engineering. Here, we review the recent progress on the biological regulatory function of LPA in bone tissue cells.
Collapse
Affiliation(s)
- Xiang-Nan Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;Hospital of Stomatology, Sun Yat-sen University, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Yuan-Yuan Ma
- Hospital of Stomatology, Sun Yat-sen University, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Zhi-Chao Hao
- Hospital of Stomatology, Sun Yat-sen University, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Hang Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Zhao X, Jiang C, Xu R, Liu Q, Liu G, Zhang Y. TRIP6 enhances stemness property of breast cancer cells through activation of Wnt/β-catenin. Cancer Cell Int 2020; 20:51. [PMID: 32082081 PMCID: PMC7023708 DOI: 10.1186/s12935-020-1136-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/04/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The urgent problem in the treatment of breast cancer is the recurrence induced by breast cancer stem cells (CSCs). Understanding the role and molecular mechanism of specific molecules in breast cancer stem cells can provide a theoretical basis for better treatment. TRIP6 is an adapter protein which belongs to the zyxin family of LIM proteins and is important in regulating the functions of CSCs. The present study aims to investigate the effects and mechanism of TRIP6 in breast cancer. METHODS TRIP6 expression in breast cancer cells and tissues were detected by Real-Time PCR, western blot and immunohistochemistry (IHC). MTT assays, colony formation assays, Xenografted tumor model and mammosphere formation assays were performed to investigate the oncogenic functions of TRIP6 in the tumorigenic capability and the tumor-initiating cell-like phenotype of breast cancer cells in vitro and in vivo. Luciferase reporter, subcellular fractionation and immunofluorescence staining assays were performed to determine the underlying mechanism of TRIP6-mediated stemness of breast cancer cells. RESULTS TRIP6 expression was significantly upregulated in breast cancer, and was closely related to the clinicopathologic characteristics, poor overall survival (OS), relapse-free survival (RFS) and poor prognosis of breast cancer patients. Functional studies revealed that overexpression of TRIP6 significantly enhanced proliferative, tumorigenicity capability and the cancer stem cell-like properties of breast cancer in vitro and in vivo. On the contrary, silencing TRIP6 achieved the opposite results. Notably, we found that TRIP6 promoted Wnt/β-catenin signaling pathway in breast cancer to strengthen the tumor-initiating cell-like phenotype of breast cancer cells. CONCLUSIONS This study indicates that TRIP6 plays an important role in maintaining the stem cell-like characteristics of breast cancer cells, supporting the significance of TRIP6 as a novel potential prognostic biomarker and therapeutic target for diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Xiaohui Zhao
- GMU-Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436 China
| | - Chao Jiang
- Department of Cancer Center, People’s Hospital of Baoan District, Shenzhen, 518101 China
| | - Rui Xu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095 China
| | - Qingnan Liu
- GMU-Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436 China
| | - Guanglin Liu
- Novartis Oncology (China) AG, Guangzhou, 510630 China
- The First Affiliation Hospital of Guangzhou Medical University, Guangzhou, 510120 China
| | - Yan Zhang
- Department of Medicine Oncology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655 Guangdong China
| |
Collapse
|
6
|
Hamyeh M, Bernex F, Larive RM, Naldi A, Urbach S, Simony-Lafontaine J, Puech C, Bakhache W, Solassol J, Coopman PJ, Hendriks WJ, Freiss G. PTPN13 induces cell junction stabilization and inhibits mammary tumor invasiveness. Am J Cancer Res 2020; 10:1016-1032. [PMID: 31938048 PMCID: PMC6956795 DOI: 10.7150/thno.38537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/03/2019] [Indexed: 02/06/2023] Open
Abstract
Clinical data suggest that the protein tyrosine phosphatase PTPN13 exerts an anti-oncogenic effect. Its exact role in tumorigenesis remains, however, unclear due to its negative impact on FAS receptor-induced apoptosis. Methods: We crossed transgenic mice deleted for PTPN13 phosphatase activity with mice that overexpress human HER2 to assess the exact role of PTPN13 in tumor development and aggressiveness. To determine the molecular mechanism underlying the PTPN13 tumor suppressor activity we developed isogenic clones of the aggressive human breast cancer cell line MDA-MB-231 overexpressing either wild type or a catalytically-inactive mutant PTPN13 and subjected these to phosphoproteomic and gene ontology analyses. We investigated the PTPN13 consequences on cell aggressiveness using wound healing and Boyden chamber assays, on intercellular adhesion using videomicroscopy, cell aggregation assay and immunofluorescence. Results: The development, growth and invasiveness of breast tumors were strongly increased by deletion of the PTPN13 phosphatase activity in transgenic mice. We observed that PTPN13 phosphatase activity is required to inhibit cell motility and invasion in the MDA-MB-231 cell line overexpressing PTPN13. In vivo, the negative PTPN13 effect on tumor invasiveness was associated with a mesenchymal-to-epithelial transition phenotype in athymic mice xenografted with PTPN13-overexpressing MDA-MB-231 cells, as well as in HER2-overexpressing mice with wild type PTPN13, compared to HER2-overexpressing mice that lack PTPN13 phosphatase activity. Phosphoproteomic and gene ontology analyses indicated a role of PTPN13 in the regulation of intercellular junction-related proteins. Finally, protein localization studies in MDA-MB-231 cells and HER2-overexpressing mice tumors confirmed that PTPN13 stabilizes intercellular adhesion and promotes desmosome formation. Conclusions: These data provide the first evidence for the negative role of PTPN13 in breast tumor invasiveness and highlight its involvement in cell junction stabilization.
Collapse
|
7
|
Suckau O, Gross I, Schrötter S, Yang F, Luo J, Wree A, Chun J, Baska D, Baumgart J, Kano K, Aoki J, Bräuer AU. LPA 1 , LPA 2 , LPA 4 , and LPA 6 receptor expression during mouse brain development. Dev Dyn 2019; 248:375-395. [PMID: 30847983 PMCID: PMC6593976 DOI: 10.1002/dvdy.23] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/17/2022] Open
Abstract
Background LPA is a small bioactive phospholipid that acts as an extracellular signaling molecule and is involved in cellular processes, including cell proliferation, migration, and differentiation. LPA acts by binding and activating at least six known G protein–coupled receptors: LPA1–6. In recent years, LPA has been suggested to play an important role both in normal neuronal development and under pathological conditions in the nervous system. Results We show the expression pattern of LPA receptors during mouse brain development by using qRT‐PCR, in situ hybridization, and immunocytochemistry. Only LPA1, LPA2,LPA4, and LPA6 mRNA transcripts were detected throughout development stages from embryonic day 16 until postnatal day 30 of hippocampus, neocortex, cerebellum, and bulbus olfactorius in our experiments, while expression of LPA3 and LPA5 genes was below detection level. In addition to our qRT‐PCR results, we also analyzed the cellular protein expression of endogenous LPA receptors, with focus on LPA1 and LPA2 within postnatal brain slices and primary neuron differentiation with and without cytoskeleton stabilization and destabilization. Conclusions The expression of LPA receptors changes depends on the developmental stage in mouse brain and in cultured hippocampal primary neurons. Interestingly, we found that commercially available antibodies for LPA receptors are largely unspecific. LPA1, ‐2, ‐4, and ‐6 genes are dynamically expressed during postnatal brain development. LPA1, ‐2, ‐4, and ‐6 genes are differently expressed in the hippocampus, neocortex, cerebellum, and bulbus olfactorius. LPA1 and ‐2 gene expression alters during neuronal differentiation. LPA1, ‐2, ‐3, ‐4, and ‐6 genes are expressed in glia cells, but differed in gene expression levels.
Collapse
Affiliation(s)
- Olga Suckau
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Isabel Gross
- Institute of Anatomy, Universitätsmedizin Rostock, Rostock, Germany.,Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Sandra Schrötter
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Fan Yang
- Albrecht Kossel Institute for Neuroregeneration, Rostock University Medical Center, Rostock, Germany
| | - Jiankai Luo
- Albrecht Kossel Institute for Neuroregeneration, Rostock University Medical Center, Rostock, Germany
| | - Andreas Wree
- Institute of Anatomy, Universitätsmedizin Rostock, Rostock, Germany
| | - Jerold Chun
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California
| | - David Baska
- Translational Animal Research Center, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Jan Baumgart
- Translational Animal Research Center, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kuniyuki Kano
- Graduate School of Pharmaceutical Science, University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Graduate School of Pharmaceutical Science, University of Tokyo, Tokyo, Japan
| | - Anja U Bräuer
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Institute of Anatomy, Universitätsmedizin Rostock, Rostock, Germany.,Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
8
|
Autotaxin-Lysophosphatidic Acid: From Inflammation to Cancer Development. Mediators Inflamm 2017; 2017:9173090. [PMID: 29430083 PMCID: PMC5753009 DOI: 10.1155/2017/9173090] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/22/2017] [Indexed: 12/13/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a ubiquitous lysophospholipid and one of the main membrane-derived lipid signaling molecules. LPA acts as an autocrine/paracrine messenger through at least six G protein-coupled receptors (GPCRs), known as LPA1–6, to induce various cellular processes including wound healing, differentiation, proliferation, migration, and survival. LPA receptors and autotaxin (ATX), a secreted phosphodiesterase that produces this phospholipid, are overexpressed in many cancers and impact several features of the disease, including cancer-related inflammation, development, and progression. Many ongoing studies aim to understand ATX-LPA axis signaling in cancer and its potential as a therapeutic target. In this review, we discuss the evidence linking LPA signaling to cancer-related inflammation and its impact on cancer progression.
Collapse
|
9
|
Knowlden SA, Hillman SE, Chapman TJ, Patil R, Miller DD, Tigyi G, Georas SN. Novel Inhibitory Effect of a Lysophosphatidic Acid 2 Agonist on Allergen-Driven Airway Inflammation. Am J Respir Cell Mol Biol 2016; 54:402-9. [PMID: 26248018 DOI: 10.1165/rcmb.2015-0124oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a pleiotropic lipid signaling molecule associated with asthma pathobiology. LPA elicits its effects by binding to at least six known cell surface G protein-coupled receptors (LPA1-6) that are expressed in the lung in a cell type-specific manner. LPA2 in particular has emerged as an attractive therapeutic target in asthma because it appears to transduce inhibitory or cell-protective signals. We studied a novel and specific small molecule LPA2 agonist (2-[4-(1,3-dioxo-1H,3H-benzoisoquinolin-2-yl)butylsulfamoyl] benzoic acid [DBIBB]) in a mouse model of house dust mite-induced allergic airway inflammation. Mice injected with DBIBB developed significantly less airway and lung inflammation compared with vehicle-treated controls. Levels of lung Th2 cytokines were also significantly attenuated by DBIBB. We conclude that pharmacologic activation of LPA2 attenuates Th2-driven allergic airway inflammation in a mouse model of asthma. Targeting LPA receptor signaling holds therapeutic promise in allergic asthma.
Collapse
Affiliation(s)
- Sara A Knowlden
- 1 Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Sara E Hillman
- 2 Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Timothy J Chapman
- 2 Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Renukadevi Patil
- 3 Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee; and.,4 Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Duane D Miller
- 4 Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Gabor Tigyi
- 3 Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee; and
| | - Steve N Georas
- 1 Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York.,2 Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
10
|
Yamana S, Tokiyama A, Mizutani K, Hirata KI, Takai Y, Rikitake Y. The Cell Adhesion Molecule Necl-4/CADM4 Serves as a Novel Regulator for Contact Inhibition of Cell Movement and Proliferation. PLoS One 2015; 10:e0124259. [PMID: 25893857 PMCID: PMC4404054 DOI: 10.1371/journal.pone.0124259] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/11/2015] [Indexed: 01/16/2023] Open
Abstract
Contact inhibition of cell movement and proliferation is critical for proper organogenesis and tissue remodeling. We show here a novel regulatory mechanism for this contact inhibition using cultured vascular endothelial cells. When the cells were confluently cultured, Necl-4 was up-regulated and localized at cell–cell contact sites where it cis-interacted with the vascular endothelial growth factor (VEGF) receptor. This interaction inhibited the tyrosine-phosphorylation of the VEGF receptor through protein-tyrosine phosphatase, non-receptor type 13 (PTPN13), eventually reducing cell movement and proliferation. When the cells were sparsely cultured, Necl-4 was down-regulated but accumulated at leading edges where it inhibited the activation of Rho-associated protein kinase through PTPN13, eventually facilitating the VEGF-induced activation of Rac1 and enhancing cell movement. Necl-4 further facilitated the activation of extracellular signal-regulated kinase 1/2, eventually enhancing cell proliferation. Thus, Necl-4 serves as a novel regulator for contact inhibition of cell movement and proliferation cooperatively with the VEGF receptor and PTPN13.
Collapse
Affiliation(s)
- Shota Yamana
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Amina Tokiyama
- Division of Signal Transduction, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Kiyohito Mizutani
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Ken-ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yoshimi Takai
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yoshiyuki Rikitake
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Signal Transduction, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
- * E-mail:
| |
Collapse
|
11
|
Kihara Y, Maceyka M, Spiegel S, Chun J. Lysophospholipid receptor nomenclature review: IUPHAR Review 8. Br J Pharmacol 2014; 171:3575-94. [PMID: 24602016 PMCID: PMC4128058 DOI: 10.1111/bph.12678] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 02/03/2014] [Accepted: 02/12/2014] [Indexed: 12/11/2022] Open
Abstract
Lysophospholipids encompass a diverse range of small, membrane-derived phospholipids that act as extracellular signals. The signalling properties are mediated by 7-transmembrane GPCRs, constituent members of which have continued to be identified after their initial discovery in the mid-1990s. Here we briefly review this class of receptors, with a particular emphasis on their protein and gene nomenclatures that reflect their cognate ligands. There are six lysophospholipid receptors that interact with lysophosphatidic acid (LPA): protein names LPA1 - LPA6 and italicized gene names LPAR1-LPAR6 (human) and Lpar1-Lpar6 (non-human). There are five sphingosine 1-phosphate (S1P) receptors: protein names S1P1 -S1P5 and italicized gene names S1PR1-S1PR5 (human) and S1pr1-S1pr5 (non-human). Recent additions to the lysophospholipid receptor family have resulted in the proposed names for a lysophosphatidyl inositol (LPI) receptor - protein name LPI1 and gene name LPIR1 (human) and Lpir1 (non-human) - and three lysophosphatidyl serine receptors - protein names LyPS1 , LyPS2 , LyPS3 and gene names LYPSR1-LYPSR3 (human) and Lypsr1-Lypsr3 (non-human) along with a variant form that does not appear to exist in humans that is provisionally named LyPS2L . This nomenclature incorporates previous recommendations from the International Union of Basic and Clinical Pharmacology, the Human Genome Organization, the Gene Nomenclature Committee, and the Mouse Genome Informatix.
Collapse
Affiliation(s)
- Yasuyuki Kihara
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research InstituteLa Jolla, CA, USA
| | - Michael Maceyka
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, School of Medicine, Virginia Commonwealth UniversityRichmond, VA, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, School of Medicine, Virginia Commonwealth UniversityRichmond, VA, USA
| | - Jerold Chun
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research InstituteLa Jolla, CA, USA
| |
Collapse
|
12
|
Lai YJ, Li MY, Yang CY, Huang KH, Tsai JC, Wang TW. TRIP6 regulates neural stem cell maintenance in the postnatal mammalian subventricular zone. Dev Dyn 2014; 243:1130-42. [DOI: 10.1002/dvdy.24161] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 06/25/2014] [Accepted: 06/25/2014] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yun-Ju Lai
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Ming-Yang Li
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Cheng-Yao Yang
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Kao-Hua Huang
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Jui-Cheng Tsai
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
| | - Tsu-Wei Wang
- Department of Life Science; National Taiwan Normal University; Taipei Taiwan
- Brain Research Center; National Yang-Ming University; Taipei Taiwan
| |
Collapse
|
13
|
Yung YC, Stoddard NC, Chun J. LPA receptor signaling: pharmacology, physiology, and pathophysiology. J Lipid Res 2014; 55:1192-214. [PMID: 24643338 DOI: 10.1194/jlr.r046458] [Citation(s) in RCA: 528] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Indexed: 12/18/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a small ubiquitous lipid found in vertebrate and nonvertebrate organisms that mediates diverse biological actions and demonstrates medicinal relevance. LPA's functional roles are driven by extracellular signaling through at least six 7-transmembrane G protein-coupled receptors. These receptors are named LPA1-6 and signal through numerous effector pathways activated by heterotrimeric G proteins, including Gi/o, G12/13, Gq, and Gs LPA receptor-mediated effects have been described in numerous cell types and model systems, both in vitro and in vivo, through gain- and loss-of-function studies. These studies have revealed physiological and pathophysiological influences on virtually every organ system and developmental stage of an organism. These include the nervous, cardiovascular, reproductive, and pulmonary systems. Disturbances in normal LPA signaling may contribute to a range of diseases, including neurodevelopmental and neuropsychiatric disorders, pain, cardiovascular disease, bone disorders, fibrosis, cancer, infertility, and obesity. These studies underscore the potential of LPA receptor subtypes and related signaling mechanisms to provide novel therapeutic targets.
Collapse
Affiliation(s)
- Yun C Yung
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| | - Nicole C Stoddard
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037 Biomedical Sciences Graduate Program, University of California, San Diego School of Medicine, La Jolla, CA 92037
| | - Jerold Chun
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
14
|
Fei J, Li J, Shen S, Zhou W. Characterization of TRIP6-dependent nasopharyngeal cancer cell migration. Tumour Biol 2013; 34:2329-35. [PMID: 23576104 DOI: 10.1007/s13277-013-0780-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 03/26/2013] [Indexed: 11/30/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a leading malignancy most often reported in endemic areas such as in Southeast Asia and the Mediterranean area. NPC remains as a major challenge for clinical management largely due to its high propensity for cancer invasion, metastasis, and recurrence. Therefore, control of NPC cell motility stands as a major obstacle for successful NPC management. The current study sought to identify a new regulator for NPC cell motility in light of previous data showing a similar role of thyroid receptor interactor protein 6 (TRIP6) in other cancer cell types. Results showed that TRIP6 is up-regulated in NPC cells as compared to normal nasopharyngeal epithelial cells. Moreover, TRIP6 overexpression/knockdown results in significant enhancement/inhibition of NPC cell migration, respectively. Interestingly, data also suggested that TRIP6 Y55E (tyrosine 55 to glutamic acid) mutant can promote cell migration more efficiently than wild type does, while Y55A (tyrosine 55 to alanine) mutant has no effects on cell migration as demonstrated with different methodology. Consistently, we also found that c-Src physically interacts with TRIP6, which suggests its potential role as a TRIP6 kinase. Taken together, these data suggested that TRIP6 is involved in the regulation of NPC cell motility, and phosphorylation of tyrosine 55 residue plays an important regulatory role for this event. These data highlight the importance of TRIP6 as a novel regulator of NPC cell motility, which warrants a good basis for further investigation on the underlying mechanism by which TRIP6 exerts this effect and the pathophysiological role TRIP6 plays in vivo.
Collapse
Affiliation(s)
- Jie Fei
- Department of Otolaryngology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, 214000, China
| | | | | | | |
Collapse
|
15
|
Kiss GN, Fells JI, Gupte R, Lee SC, Liu J, Nusser N, Lim KG, Ray RM, Lin FT, Parrill AL, Sümegi B, Miller DD, Tigyi G. Virtual screening for LPA2-specific agonists identifies a nonlipid compound with antiapoptotic actions. Mol Pharmacol 2012; 82:1162-73. [PMID: 22968304 DOI: 10.1124/mol.112.079699] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Lysophosphatidic acid (LPA) is a highly potent endogenous lipid mediator that protects and rescues cells from programmed cell death. Earlier work identified the LPA₂ G protein-coupled receptor subtype as an important molecular target of LPA mediating antiapoptotic signaling. Here we describe the results of a virtual screen using single-reference similarity searching that yielded compounds 2-((9-oxo-9H-fluoren-2-yl)carbamoyl)benzoic acid (NSC12404), 2-((3-(1,3-dioxo-1H-benzo[de]isoquinolin-2(3H)-yl)propyl)thio)benzoic acid (GRI977143), 4,5-dichloro-2-((9-oxo-9H-fluoren-2-yl)carbamoyl)benzoic acid (H2L5547924), and 2-((9,10-dioxo-9,10-dihydroanthracen-2-yl)carbamoyl) benzoic acid (H2L5828102), novel nonlipid and drug-like compounds that are specific for the LPA₂ receptor subtype. We characterized the antiapoptotic action of one of these compounds, GRI977143, which was effective in reducing activation of caspases 3, 7, 8, and 9 and inhibited poly(ADP-ribose)polymerase 1 cleavage and DNA fragmentation in different extrinsic and intrinsic models of apoptosis in vitro. Furthermore, GRI977143 promoted carcinoma cell invasion of human umbilical vein endothelial cell monolayers and fibroblast proliferation. The antiapoptotic cellular signaling responses were present selectively in mouse embryonic fibroblast cells derived from LPA(1&2) double-knockout mice reconstituted with the LPA₂ receptor and were absent in vector-transduced control cells. GRI977143 was an effective stimulator of extracellular signal-regulated kinase 1/2 activation and promoted the assembly of a macromolecular signaling complex consisting of LPA₂, Na⁺ - H⁺ exchange regulatory factor 2, and thyroid receptor interacting protein 6, which has been shown previously to be a required step in LPA-induced antiapoptotic signaling. The present findings indicate that nonlipid LPA₂-specific agonists represent an excellent starting point for development of lead compounds with potential therapeutic utility for preventing the programmed cell death involved in many types of degenerative and inflammatory diseases.
Collapse
Affiliation(s)
- Gyöngyi N Kiss
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
Integrating signals from the ECM (extracellular matrix) via the cell surface into the nucleus is an essential feature of multicellular life and often malfunctions in cancer. To date many signal transducers known as shuttle proteins have been identified that act as both: a cytoskeletal and a signalling protein. Here, we highlight the interesting member of the Zyxin family TRIP6 [thyroid receptor interactor protein 6; also designated ZRP-1 (zyxin-related protein 1)] and review current literature to define its role in cell physiology and cancer. TRIP6 is a versatile scaffolding protein at FAs (focal adhesions) involved in cytoskeletal organization, coordinated cell migration and tissue invasion. Via its LIM and TDC domains TRIP6 interacts with different components of the LPA (lysophosphatidic acid), NF-κB (nuclear factor κB), glucocorticoid and AMPK (AMP-activated protein kinase) signalling pathway and thereby modulates their activity. Within the nucleus TRIP6 acts as a transcriptional cofactor regulating the transcriptional responses of these pathways. Moreover, intranuclear TRIP6 associates with proteins ensuring telomere protection and hence may contribute to genome stability. Accordingly, TRIP6 is engaged in key cellular processes such as cell proliferation, differentiation and survival. These diverse functions of TRIP6 are found to be dysregulated in various cancers and may have pleiotropic roles in tumour initiation, tumour growth and metastasis, which turn TRIP6 into an attractive candidate for cancer diagnosis and targeted therapy.
Collapse
|
17
|
Castilla C, Flores ML, Conde JM, Medina R, Torrubia FJ, Japón MA, Sáez C. Downregulation of protein tyrosine phosphatase PTPL1 alters cell cycle and upregulates invasion-related genes in prostate cancer cells. Clin Exp Metastasis 2012; 29:349-58. [PMID: 22274591 DOI: 10.1007/s10585-012-9455-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 01/13/2012] [Indexed: 12/30/2022]
Abstract
PTPL1, a non-receptor type protein tyrosine phosphatase, has been involved in the regulation of apoptosis and invasiveness of various tumour cell types, but its role in prostate cancer remained to be investigated. We report here that downregulation of PTPL1 by small interfering RNA in PC3 cells decreases cell proliferation and concomitantly reduces the expression of cell cycle-related proteins such as cyclins E and B1, PCNA, PTTG1 and phospho-histone H3. PTPL1 downregulation also increases the invasion ability of PC3 cells through Matrigel coated membranes. cDNA array of PTPL1-silenced PC3 cells versus control cells showed an upregulation of invasion-related genes such as uPA, uPAR, tPA, PAI-1, integrin α6 and osteopontin. This increased expression was also confirmed in PTPL1-silenced DU145 prostate cancer cells by quantitative real time PCR and western blot. These findings suggest that PTPL1 is an important mediator of central cellular processes such as proliferation and invasion.
Collapse
Affiliation(s)
- Carolina Castilla
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avenida Manuel Siurot s/n, 41013 Seville, Spain
| | | | | | | | | | | | | |
Collapse
|
18
|
Lin VTG, Lin FT. TRIP6: an adaptor protein that regulates cell motility, antiapoptotic signaling and transcriptional activity. Cell Signal 2011; 23:1691-7. [PMID: 21689746 PMCID: PMC3156290 DOI: 10.1016/j.cellsig.2011.06.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 06/01/2011] [Accepted: 06/06/2011] [Indexed: 01/02/2023]
Abstract
Thyroid hormone receptor interacting protein 6 (TRIP6), also known as zyxin-related protein-1 (ZRP-1), is an adaptor protein that belongs to the zyxin family of LIM proteins. TRIP6 is primarily localized in the cytosol or focal adhesion plaques, and may associate with the actin cytoskeleton. Additionally, it is capable of shuttling to the nucleus to serve as a transcriptional coregulator. Structural and functional analyses have revealed that through multidomain-mediated protein-protein interactions, TRIP6 serves as a platform for the recruitment of a wide variety of signaling molecules involved in diverse cellular responses, such as actin cytoskeletal reorganization, cell adhesion and migration, antiapoptotic signaling, osteoclast sealing zone formation and transcriptional control. Although the physiological functions of TRIP6 remain largely unknown, it has been implicated in cancer progression and telomere protection. Together, these studies suggest that TRIP6 plays multifunctional roles in different cellular responses, and thus may represent a novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Victor T. G. Lin
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294-0005
| | - Fang-Tsyr Lin
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294-0005
| |
Collapse
|
19
|
Abstract
Over 250 PDZ (PSD95/Dlg/ZO-1) domain-containing proteins have been described in the human proteome. As many of these possess multiple PDZ domains, the potential combinations of associations with proteins that possess PBMs (PDZ-binding motifs) are vast. However, PDZ domain recognition is a highly specific process, and much less promiscuous than originally thought. Furthermore, a large number of PDZ domain-containing proteins have been linked directly to the control of processes whose loss, or inappropriate activation, contribute to the development of human malignancies. These regulate processes as diverse as cytoskeletal organization, cell polarity, cell proliferation and many signal transduction pathways. In the present review, we discuss how PBM–PDZ recognition and imbalances therein can perturb cellular homoeostasis and ultimately contribute to malignant progression.
Collapse
|
20
|
The adaptor protein TRIP6 antagonizes Fas-induced apoptosis but promotes its effect on cell migration. Mol Cell Biol 2010; 30:5582-96. [PMID: 20876301 DOI: 10.1128/mcb.00134-10] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The Fas/CD95 receptor mediates apoptosis but is also capable of triggering nonapoptotic signals. However, the mechanisms that selectively regulate these opposing effects are not yet fully understood. Here we demonstrate that the activation of Fas or stimulation with lysophosphatidic acid (LPA) induces cytoskeletal reorganization, leading to the association of Fas with actin stress fibers and the adaptor protein TRIP6. TRIP6 binds to the cytoplasmic juxtamembrane domain of Fas and interferes with the recruitment of FADD to Fas. Furthermore, through physical interactions with NF-κB p65, TRIP6 regulates nuclear translocation and the activation of NF-κB upon Fas activation or LPA stimulation. As a result, TRIP6 antagonizes Fas-induced apoptosis and further enhances the antiapoptotic effect of LPA in cells that express high levels of TRIP6. On the other hand, TRIP6 promotes Fas-mediated cell migration in apoptosis-resistant glioma cells. This effect is regulated via the Src-dependent phosphorylation of TRIP6 at Tyr-55. As TRIP6 is overexpressed in glioblastomas, this may have a significant impact on enhanced NF-κB activity, resistance to apoptosis, and Fas-mediated cell invasion in glioblastomas.
Collapse
|
21
|
Glondu-Lassis M, Dromard M, Lacroix-Triki M, Nirdé P, Puech C, Knani D, Chalbos D, Freiss G. PTPL1/PTPN13 regulates breast cancer cell aggressiveness through direct inactivation of Src kinase. Cancer Res 2010; 70:5116-26. [PMID: 20501847 DOI: 10.1158/0008-5472.can-09-4368] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The protein tyrosine phosphatase PTPL1/PTPN13, the activity of which is decreased through allelic loss, promoter methylation, or somatic mutations in some tumors, has been proposed as a tumor suppressor gene. Moreover, our recent clinical study identified PTPL1 expression level as an independent prognostic indicator of a favorable outcome for patients with breast cancer. However, how PTPL1 can affect tumor aggressiveness has not been characterized. Here, we first show that PTPL1 expression, assessed by immunohistochemistry, is decreased in breast cancer and metastasis specimens compared with nonmalignant tissues. Second, to evaluate whether PTPL1 plays a critical role in breast cancer progression, RNA interference experiments were performed in poorly tumorigenic MCF-7 breast cancer cells. PTPL1 inhibition drastically increased tumor growth in athymic mice and also enhanced several parameters associated with tumor progression, including cell proliferation on extracellular matrix components and cell invasion. Furthermore, the inhibition of Src kinase expression drastically blocked the effects of PTPL1 silencing on cell growth. In PTPL1 knockdown cells, the phosphorylation of Src on tyrosine 419 is increased, leading to the activation of its downstream substrates Fak and p130cas. Finally, substrate-trapping experiments revealed that Src tyrosine 419 is a direct target of the phosphatase. Thus, by identification of PTPL1 as the first phosphatase able to inhibit Src through direct dephosphorylation in intact cells, we presently describe a new mechanism by which PTPL1 inhibits breast tumor aggressiveness.
Collapse
|
22
|
Choi JW, Herr DR, Noguchi K, Yung YC, Lee CW, Mutoh T, Lin ME, Teo ST, Park KE, Mosley AN, Chun J. LPA receptors: subtypes and biological actions. Annu Rev Pharmacol Toxicol 2010; 50:157-86. [PMID: 20055701 DOI: 10.1146/annurev.pharmtox.010909.105753] [Citation(s) in RCA: 649] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lysophosphatidic acid (LPA) is a small, ubiquitous phospholipid that acts as an extracellular signaling molecule by binding to and activating at least five known G protein-coupled receptors (GPCRs): LPA(1)-LPA(5). They are encoded by distinct genes named LPAR1-LPAR5 in humans and Lpar1-Lpar5 in mice. The biological roles of LPA are diverse and include developmental, physiological, and pathophysiological effects. This diversity is mediated by broad and overlapping expression patterns and multiple downstream signaling pathways activated by cognate LPA receptors. Studies using cloned receptors and genetic knockout mice have been instrumental in uncovering the significance of this signaling system, notably involving basic cellular processes as well as multiple organ systems such as the nervous system. This has further provided valuable proof-of-concept data to support LPA receptors and LPA metabolic enzymes as targets for the treatment of medically important diseases that include neuropsychiatric disorders, neuropathic pain, infertility, cardiovascular disease, inflammation, fibrosis, and cancer.
Collapse
Affiliation(s)
- Ji Woong Choi
- Department of Molecular Biology, Helen L. Dorris Institute for Neurological and Psychiatric Disorders, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Stenzel N, Fetzer CP, Heumann R, Erdmann KS. PDZ-domain-directed basolateral targeting of the peripheral membrane protein FRMPD2 in epithelial cells. J Cell Sci 2009; 122:3374-84. [PMID: 19706687 DOI: 10.1242/jcs.046854] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Multi-PDZ (PSD-95/Discs large/Zonula-occludens-1) domain proteins play a crucial role in the establishment and maintenance of cell polarization. The novel multi-PDZ domain protein FRMPD2 is a potential scaffolding protein consisting of an N-terminal KIND domain, a FERM domain and three PDZ domains. Here we show that FRMPD2 is localized in a polarized fashion in epithelial cells at the basolateral membrane and partially colocalizes with the tight-junction marker protein Zonula-occludens-1. Downregulation of FRMPD2 protein in Caco-2 cells is associated with an impairment of tight junction formation. We find that the FERM domain of FRMPD2 binds phosphatidylinositols and is sufficient for membrane localization. Moreover, we demonstrate that recruitment of FRMPD2 to cell-cell junctions is strictly E-cadherin-dependent, which is in line with our identification of catenin family proteins as binding partners for FRMPD2. We demonstrate that the FERM domain and binding of the PDZ2 domain to the armadillo protein p0071 are required for basolateral restriction of FRMPD2. Moreover, the PDZ2 domain of FRMPD2 is sufficient to partially redirect an apically localized protein to the basolateral membrane. Our results provide novel insights into the molecular function of FRMPD2 and into the targeting mechanism of peripheral membrane proteins in polarized epithelial cells.
Collapse
Affiliation(s)
- Nina Stenzel
- Department of Biochemistry II, Ruhr-University Bochum, 44780 Bochum, Germany
| | | | | | | |
Collapse
|
24
|
Crowley JL, Smith TC, Fang Z, Takizawa N, Luna EJ. Supervillin reorganizes the actin cytoskeleton and increases invadopodial efficiency. Mol Biol Cell 2008; 20:948-62. [PMID: 19109420 DOI: 10.1091/mbc.e08-08-0867] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Tumor cells use actin-rich protrusions called invadopodia to degrade extracellular matrix (ECM) and invade tissues; related structures, termed podosomes, are sites of dynamic ECM interaction. We show here that supervillin (SV), a peripheral membrane protein that binds F-actin and myosin II, reorganizes the actin cytoskeleton and potentiates invadopodial function. Overexpressed SV induces redistribution of lamellipodial cortactin and lamellipodin/RAPH1/PREL1 away from the cell periphery to internal sites and concomitantly increases the numbers of F-actin punctae. Most punctae are highly dynamic and colocalize with the podosome/invadopodial proteins, cortactin, Tks5, and cdc42. Cortactin binds SV sequences in vitro and contributes to the formation of enhanced green fluorescent protein (EGFP)-SV induced punctae. SV localizes to the cores of Src-generated podosomes in COS-7 cells and with invadopodia in MDA-MB-231 cells. EGFP-SV overexpression increases average numbers of ECM holes per cell; RNA interference-mediated knockdown of SV decreases these numbers. Although SV knockdown alone has no effect, simultaneous down-regulation of SV and the closely related protein gelsolin reduces invasion through ECM. Together, our results show that SV is a component of podosomes and invadopodia and that SV plays a role in invadopodial function, perhaps as a mediator of cortactin localization, activation state, and/or dynamics of metalloproteinases at the ventral cell surface.
Collapse
Affiliation(s)
- Jessica L Crowley
- Department of Cell Biology and Cell Dynamics Program, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | |
Collapse
|
25
|
Abstract
Protein tyrosine phosphatase, PTPL1, (also known as PTPN13, FAP-1, PTP-BAS, PTP1E) is a non-receptor type PTP and, at 270 kDa, is the largest phosphatase within this group. In addition to the well-conserved PTP domain, PTPL1 contains at least 7 putative macromolecular interaction domains. This structural complexity indicates that PTPL1 may modulate diverse cellular functions, perhaps exerting both positive and negative effects. In accordance with this idea, while certain studies suggest that PTPL1 can act as a tumor-promoting gene other experimental studies have suggested that PTPL1 may function as a tumor suppressor. The role of PTPL1 in the cancer cell is therefore likely to be both complex and context dependent with possible roles including the modulation of growth, stress-response, and cytoskeletal remodeling pathways. Understanding the nature of molecular complexes containing PTPL1, its interaction partners, substrates, regulation and subcellular localization are key to unraveling the complex personality of this protein phosphatase.
Collapse
|
26
|
Lin FT, Lai YJ. Regulation of the LPA2 receptor signaling through the carboxyl-terminal tail-mediated protein-protein interactions. Biochim Biophys Acta Mol Cell Biol Lipids 2008; 1781:558-62. [PMID: 18501721 DOI: 10.1016/j.bbalip.2008.04.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Revised: 04/22/2008] [Accepted: 04/22/2008] [Indexed: 01/08/2023]
Abstract
While it is well known that lysophosphatidic acid (LPA) mediates diverse physiological and pathophysiological responses through the activation of G protein-coupled LPA receptors, the specificity and molecular mechanisms by which different LPA receptors mediate these biological responses remain largely unknown. Recent identification of several PDZ proteins and zinc finger proteins that interact with the carboxyl-terminal tail of the LPA2 receptor provides a considerable progress towards the understanding of the mechanisms how the LPA2 receptor specifically mediates LPA signaling pathways. These findings have led to the proposal that there are at least two distinct protein interaction motifs present in the carboxyl-terminus of the LPA2 receptor. Together, these data provide a new concept that the efficiency and specificity of the LPA2 receptor-mediated signal transduction can be achieved through the cross-regulation between the classical G protein-activated signaling cascades and the interacting partner-mediated signaling pathways.
Collapse
Affiliation(s)
- Fang-Tsyr Lin
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294-0005, USA.
| | | |
Collapse
|