1
|
Huang W, Zhou H, He Y, Wang A, Wang B, Chen Y, Liu C, Wang H, Xie W, Kong H. A novel PDGFR inhibitor WQ-C-401 prevents pulmonary vascular remodeling in rats with monocrotaline-induced pulmonary arterial hypertension. Exp Cell Res 2024; 441:114154. [PMID: 38996959 DOI: 10.1016/j.yexcr.2024.114154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Platelet-derived growth factor (PDGF) is one of the most important cytokines associated with pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). PDGF receptor (PDGFR) inhibition exerted therapeutic effects on PAH in clinical trials, but serious side effects warrant the withdrawal of existing drugs. In this study, a novel highly selective PDGFR inhibitor WQ-C-401 was developed, and its effects on PDGFR signaling pathway and pulmonary vascular remodeling in PAH were investigated. Cell proliferation assays and Western blot analysis of PDGFRα/β phosphorylation showed that WQ-C-401 inhibited PDGFR-mediated cell proliferation assay and suppressed PDGFR phosphorylation in a concentration-dependent manner. DiscoverX's KinomeScanTM technology confirmed the good kinome selectivity of WQ-C-401 (S score (1) of PDGFR = (0.01)). In monocrotaline (MCT)-induced PAH rats, intragastric administration of WQ-C-401 (25, 50, 100 mg/kg/d) or imatinib (50 mg/kg/d, positive control) significantly decreased right ventricular systolic pressure (RVSP). Histological analysis demonstrated that WQ-C-401 inhibited pulmonary vascular remodeling by reducing muscularization and fibrosis, as well as alleviated right ventricular hypertrophy in MCT-treated rats. In addition, WQ-C-401 suppressed MCT-induced cell hyperproliferation and CD68+ macrophage infiltration around the pulmonary artery. In vitro, WQ-C-401 inhibited PDGF-BB-induced proliferation and migration of human pulmonary arterial smooth muscle cells (PASMCs). Moreover, Western blot analysis showed that WQ-C-401 concertration-dependently inhibited PDGF-BB-induced phosphorylation of ERK1/2 and PDGFRβ Y751, decreased collagen Ⅰ synthesis and increased alpha smooth muscle actin (α-SMA) expression in PASMCs. Collectively, our results suggest that WQ-C-401 is a selective and potent PDGFR inhibitor which could be a promising drug for the therapeutics of PAH by preventing pulmonary vascular remodeling.
Collapse
MESH Headings
- Animals
- Monocrotaline
- Vascular Remodeling/drug effects
- Rats
- Cell Proliferation/drug effects
- Male
- Rats, Sprague-Dawley
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/chemically induced
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/pathology
- Humans
- Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors
- Receptors, Platelet-Derived Growth Factor/metabolism
- Phosphorylation/drug effects
- Pulmonary Artery/drug effects
- Pulmonary Artery/pathology
- Pulmonary Artery/metabolism
- Signal Transduction/drug effects
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/prevention & control
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/metabolism
- Protein Kinase Inhibitors/pharmacology
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
Collapse
Affiliation(s)
- Wen Huang
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Hong Zhou
- Department of Pulmonary & Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, Jiangsu, PR China
| | - Yiting He
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Aoli Wang
- Anhui Province Key Laboratory of Medical Physics & Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China
| | - Beilei Wang
- Anhui Province Key Laboratory of Medical Physics & Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China
| | - Yongfei Chen
- Anhui Province Key Laboratory of Medical Physics & Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, Anhui, PR China
| | - Chenyang Liu
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Hong Wang
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China
| | - Weiping Xie
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China.
| | - Hui Kong
- Department of Pulmonary & Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, Jiangsu, PR China.
| |
Collapse
|
2
|
Wang H, Zhang L, Liu H, Yang Y, Lu W, Cao X, Yang X, Qin Q, Song R, Feng D, Wang S, Bai T, He J. PDZK1 confers sensitivity to sunitinib in clear cell renal cell carcinoma by suppressing the PDGFR-β pathway. Br J Cancer 2024; 131:347-360. [PMID: 38822145 PMCID: PMC11263541 DOI: 10.1038/s41416-024-02725-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Sunitinib has emerged as the primary treatment for advanced or metastatic clear cell renal cell carcinoma (ccRCC) due to its significant improvement in patients' average survival time. However, drug resistance and adverse effects of sunitinib pose challenges to its clinical benefits. METHODS The differentially expressed genes (DEGs) associated with sunitinib sensitivity and resistance in ccRCC were investigated. Cell counting kit-8, plate colony formation, flow cytometry and subcutaneous xenograft tumor model assays were employed to explore the effects of PDZK1 on ccRCC. Further research on the molecular mechanism was conducted through western blot, co-immunoprecipitation, immunofluorescence co-localization and immunohistochemical staining. RESULTS We elucidated that PDZK1 is significantly downregulated in sunitinib-resistant ccRCC specimens, and PDZK1 negatively regulates the phosphorylation of PDGFR-β and the activation of its downstream pathways through interaction with PDGFR-β. The dysregulated low levels of PDZK1 contribute to inadequate inhibition of cell proliferation, tumor growth, and insensitivity to sunitinib treatment. Notably, our preclinical investigations showed that miR-15b antagomirs enhance sunitinib cytotoxic effects against ccRCC cells by upregulating PDZK1 levels, suggesting their potential in overcoming sunitinib resistance. CONCLUSIONS Our findings establish the miR-15b/PDZK1/PDGFR-β axis as a promising therapeutic target and a novel predictor for ccRCC patients' response to sunitinib treatment.
Collapse
MESH Headings
- Sunitinib/pharmacology
- Sunitinib/therapeutic use
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/metabolism
- Humans
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/pathology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Animals
- Drug Resistance, Neoplasm/genetics
- Mice
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Xenograft Model Antitumor Assays
- MicroRNAs/genetics
- Signal Transduction/drug effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Male
- Mice, Nude
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
Collapse
Affiliation(s)
- Haibo Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Lijie Zhang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hua Liu
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Yumeng Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Wenxiu Lu
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Xuedi Cao
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
| | - Xiaomei Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Qiong Qin
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Ran Song
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Duiping Feng
- Department of Interventional Radiology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Songlin Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, People's Republic of China
- Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy and Tooth Regeneration, School of Stomatology, Capital Medical University, Beijing, People's Republic of China
| | - Tao Bai
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Junqi He
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, People's Republic of China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
3
|
van Outersterp I, van der Velden VH, Hoogeveen PG, Vaitkevičienė GE, Sonneveld E, van Haaften G, Kuiper RP, zur Stadt U, Escherich G, Boer JM, den Boer ML. ABL-class Genomic Breakpoint Q-PCR: A Patient-specific Approach for MRD Monitoring in Acute Lymphoblastic Leukemia. Hemasphere 2023; 7:e967. [PMID: 37736661 PMCID: PMC10511034 DOI: 10.1097/hs9.0000000000000967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/03/2023] [Indexed: 09/23/2023] Open
Affiliation(s)
| | - Vincent H.J. van der Velden
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Patricia G. Hoogeveen
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Goda E. Vaitkevičienė
- Faculty of Medicine, Vilnius University, Lithuania
- Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Lithuania
| | - Edwin Sonneveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Dutch Childhood Oncology Group, Utrecht, The Netherlands
| | - Gijs van Haaften
- Department of Genetics, University Medical Center Utrecht, The Netherlands
| | - Roland P. Kuiper
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Genetics, University Medical Center Utrecht, The Netherlands
| | - Udo zur Stadt
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Judith M. Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Monique L. den Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology and Hematology, Erasmus MC - Sophia Children’s Hospital, Rotterdam, The Netherlands
| |
Collapse
|
4
|
Sun T, Bi F, Liu Z, Yang Q. TMEM119 facilitates ovarian cancer cell proliferation, invasion, and migration via the PDGFRB/PI3K/AKT signaling pathway. J Transl Med 2021; 19:111. [PMID: 33731124 PMCID: PMC7968362 DOI: 10.1186/s12967-021-02781-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/08/2021] [Indexed: 12/28/2022] Open
Abstract
Background Ovarian cancer (OV) is the deadliest gynecological cancer. Transmembrane protein 119 (TMEM119) has been reported as oncogene in several human cancers. However, the function of TMEM119 in OV is still poorly known. Methods Western blot and qRT-PCR were used to analyze TMEM119 levels. Transwell assays, wound healing assays, CCK-8 assays and EdU cell proliferation assays were designed to explore the function and potential mechanism of TMEM119 in malignant biological behaviors in OV. Results TMEM119 was observed to be overexpressed in OV tissues and associated with poor survival in OV patients. Knockdown and overexpression experiments demonstrated that TMEM119 promoted proliferation, invasion, and migration in OV cells in vitro. TMEM119 mRNA expression was related to the pathways of focal adhesion according to Gene Set Enrichment Analyses and was correlated with the mRNA expression level of platelet-derived growth factor receptor beta (PDGFRB). TMEM119 exerted oncogenic effects partially by regulating the expression of PDGFRB and by activating the PI3K/AKT signaling pathway. Conclusions Collectively, our findings highlight the potential role of TMEM119 in the malignant biological behavior of OV, which may serve as a potential biomarker and a therapeutic candidate for OV. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02781-x.
Collapse
Affiliation(s)
- Tianshui Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China
| | - Fangfang Bi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China
| | - Zhuonan Liu
- Department of Urology, First Hospital of China Medical University, Shenyang, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
5
|
Xu G, Liu C, Liang T, Zhang Z, Jiang J, Chen J, Xue J, Zeng H, Lu Z, Zhan X. Gene expression profile and bioinformatics analysis revealed key molecular characteristics of chordoma-before and after TNF- a treatment. Medicine (Baltimore) 2020; 99:e18790. [PMID: 32011476 PMCID: PMC7220412 DOI: 10.1097/md.0000000000018790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 11/16/2019] [Accepted: 12/16/2019] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Chordoma is a rare malignant tumor with limited treatment. Recent studies have shown that the proliferation and invasion ability of chordoma after Tumor necrosis factor alpha (TNF-α) treatment is enhanced, which may activate the gene pathway involved in the development of chordoma. This study tends to identify differentially expressed genes (DEGs) before and after treatment of TNF-α in chordoma cell line, providing a new target for future molecular therapy of chordoma. METHODS The gene expression profile of GSE101867 was downloaded from the Gene Expression Omnibus database, and the differentially expressed genes were obtained using GEO2R. Based on the CLUEGO plugin in Cytoscape, DEGs functionality and enrichment analysis. A protein-protein interaction (PPI) network was constructed using Cytoscape based on data collected from the STRING online dataset. The Hub genes are selected from the CytoHubba, the first 20 genes that coexist with the KEGG tumor-related pathway. RESULTS A total of 560 genes, including 304 up-regulated genes and 256 down-regulated genes, were selected as DEGs. Obviously, GO analysis shows that up-regulated and down-regulated DEGs are mainly enriched in biological processes such as synaptic tissue, cell adhesion, extracellular matrix organization and skeletal system development. DEGs are mainly enriched in tumor-associated pathways such as Pi3k-akt Signal path, Rap1 signal path. Three key genes were identified: PDGFRB, KDR, FGF2. All of these genes are involved in the tumor-associated pathways described previously. CONCLUSION This study is helpful in understanding the molecular characteristics of chordoma development. Hub genes PDGFRB, KDR, FGF2 and pi3k-akt signaling pathway, Rap1 signaling pathway will become a new target for the future treatment of chordoma.
Collapse
Affiliation(s)
| | - Chong Liu
- Guangxi Medical University
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | | | | | | | | | | | | | | | - Xinli Zhan
- Guangxi Medical University
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| |
Collapse
|
6
|
Roskoski R. The role of small molecule platelet-derived growth factor receptor (PDGFR) inhibitors in the treatment of neoplastic disorders. Pharmacol Res 2018; 129:65-83. [DOI: 10.1016/j.phrs.2018.01.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 01/29/2018] [Indexed: 12/15/2022]
|
7
|
Seong J, Huang M, Sim KM, Kim H, Wang Y. FRET-based Visualization of PDGF Receptor Activation at Membrane Microdomains. Sci Rep 2017; 7:1593. [PMID: 28487538 PMCID: PMC5431615 DOI: 10.1038/s41598-017-01789-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 03/31/2017] [Indexed: 11/09/2022] Open
Abstract
Platelet-derived growth factor receptor (PDGFR) senses extracellular growth factors and transfer the signals inside the cells regulating cell proliferation, migration and survival. It has been controversial at which membrane microdomains PDGFRs reside and how they control such diverse intracellular signaling pathways. Here, we developed a novel PDGFR biosensor based on fluorescence resonance energy transfer (FRET), which can detect the real-time PDGFR activity in live cells with high spatiotemporal resolutions. To study subcellular PDGFR activity at membrane microdomains, this PDGFR biosensor was further targeted in or outside lipid rafts via different lipid modification signals. The results suggest that, in response to PDGF stimulation, PDGFR activity is evenly distributed at different membrane microdomains, while integrin-mediated signaling events have inhibitory effects on the activation of PDGFR specifically located in lipid rafts but not outside rafts, implying the role of lipid microdomains as segregated signaling platforms.
Collapse
Affiliation(s)
- Jihye Seong
- Neuroscience Program, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA. .,Convergence Research Center for Diagnosis Treatment Care of Dementia, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea. .,Biological Chemistry Program, Korea University of Science and Technology (UST), Daejeon, 34113, South Korea. .,Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, South Korea.
| | - Min Huang
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA
| | - Kyoung Mi Sim
- Convergence Research Center for Diagnosis Treatment Care of Dementia, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
| | - Hyunbin Kim
- Convergence Research Center for Diagnosis Treatment Care of Dementia, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea.,Department of Converging Science and Technology, Kyung Hee University, Seoul, 02447, South Korea
| | - Yingxiao Wang
- Neuroscience Program, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA. .,Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL, 61801, USA. .,Department of Bioengineering, University of California, San Diego, CA, 92093, USA.
| |
Collapse
|
8
|
Haan S, Bahlawane C, Wang J, Nazarov PV, Muller A, Eulenfeld R, Haan C, Rolvering C, Vallar L, Satagopam VP, Sauter T, Wiesinger MY. The oncogenic FIP1L1-PDGFRα fusion protein displays skewed signaling properties compared to its wild-type PDGFRα counterpart. JAKSTAT 2015; 4:e1062596. [PMID: 26413425 PMCID: PMC4583054 DOI: 10.1080/21623996.2015.1062596] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/05/2015] [Accepted: 06/09/2015] [Indexed: 01/05/2023] Open
Abstract
Aberrant activation of oncogenic kinases is frequently observed in human cancers, but the underlying mechanism and resulting effects on global signaling are incompletely understood. Here, we demonstrate that the oncogenic FIP1L1-PDGFRα kinase exhibits a significantly different signaling pattern compared to its PDGFRα wild type counterpart. Interestingly, the activation of primarily membrane-based signal transduction processes (such as PI3-kinase- and MAP-kinase- pathways) is remarkably shifted toward a prominent activation of STAT factors. This diverging signaling pattern compared to classical PDGF-receptor signaling is partially coupled to the aberrant cytoplasmic localization of the oncogene, since membrane targeting of FIP1L1-PDGFRα restores activation of MAPK- and PI3K-pathways. In stark contrast to the classical cytokine-induced STAT activation process, STAT activation by FIP1L1-PDGFRα does neither require Janus kinase activity nor Src kinase activity. Furthermore, we investigated the mechanism of STAT5 activation via FIP1L1-PDGFRα in more detail and found that STAT5 activation does not involve an SH2-domain-mediated binding mechanism. We thus demonstrate that STAT5 activation occurs via a non-canonical activation mechanism in which STAT5 may be subject to a direct phosphorylation by FIP1L1-PDGFRα.
Collapse
Affiliation(s)
- Serge Haan
- Molecular Disease Mechanisms Group; Life Sciences Research Unit; University of Luxembourg; Luxembourg , Luxembourg
| | - Christelle Bahlawane
- Molecular Disease Mechanisms Group; Life Sciences Research Unit; University of Luxembourg; Luxembourg , Luxembourg
| | - Jiali Wang
- Molecular Disease Mechanisms Group; Life Sciences Research Unit; University of Luxembourg; Luxembourg , Luxembourg
| | - Petr V Nazarov
- Genomics Research Unit; Luxembourg Institute of Health; Luxembourg , Luxembourg
| | - Arnaud Muller
- Genomics Research Unit; Luxembourg Institute of Health; Luxembourg , Luxembourg
| | - René Eulenfeld
- Signal Transduction Group; Life Sciences Research Unit; University of Luxembourg; Luxembourg , Luxembourg
| | - Claude Haan
- Signal Transduction Group; Life Sciences Research Unit; University of Luxembourg; Luxembourg , Luxembourg
| | - Catherine Rolvering
- Signal Transduction Group; Life Sciences Research Unit; University of Luxembourg; Luxembourg , Luxembourg
| | - Laurent Vallar
- Genomics Research Unit; Luxembourg Institute of Health; Luxembourg , Luxembourg
| | - Venkata P Satagopam
- Luxembourg Center for Systems Biomedicine; University of Luxembourg ; Esch-sur-Alzette, Luxembourg
| | - Thomas Sauter
- Systems Biology Group; Life Sciences Research Unit; University of Luxembourg; Luxembourg , Luxembourg
| | - Monique Yvonne Wiesinger
- Systems Biology Group; Life Sciences Research Unit; University of Luxembourg; Luxembourg , Luxembourg
| |
Collapse
|
9
|
Yu J, Rupasinghe C, Wilson JL, Taylor L, Rahimi N, Mierke D, Polgar P. Targeting receptor tyrosine kinases and their downstream signaling with cell-penetrating peptides in human pulmonary artery smooth muscle and endothelial cells. Chem Biol Drug Des 2015; 85:586-97. [PMID: 25298192 PMCID: PMC4390552 DOI: 10.1111/cbdd.12446] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/12/2014] [Accepted: 09/25/2014] [Indexed: 01/05/2023]
Abstract
Cell-penetrating peptide (CPP) intracellular delivery of receptor signaling motifs provides an opportunity to regulate specific receptor tyrosine kinase signal transductions. We targeted tyrosine residues Y740 and Y751 of the PDGF receptor β (PDGFRβ) and Y1175 of the VEGF receptor 2 (VEGFR2). The Y740 and Y751 motifs activated ERK and Akt, while the Y1175 motif activated ERK. Targeting either Y740 or Y751 of the PDGFRβ in human pulmonary artery smooth muscle cells (HPASMC) effectively inhibited PDGF activation of ERK or Akt. Interfering with the Y751 region of the PDGFRβ proved more effective than targeting the Y740 region. The phosphorylation of Y751 of the CPP and the length and exact sequence of the mimicking peptide proved crucial. On the other hand, in human pulmonary artery endothelial cell phosphorylation of the VEGFR2 Y1175 CPP was not a determinant in blockage of ERK activation. Likewise, the length of the peptide mimic was not crucial with a very small sequence containing the Y1175 remaining effective. Physiologic proof of concept for the effectiveness of the CPP was confirmed by blockage of HPASMC migration in response to PDGF following culture injury. Thus targeted blockage of tyrosine kinase receptor signaling can be very effective.
Collapse
Affiliation(s)
- Jun Yu
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | | | - Jamie L. Wilson
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Linda Taylor
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Nader Rahimi
- Departments of Pathology and Lab Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Dale Mierke
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Peter Polgar
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
10
|
Bedekovics J, Kiss A, Beke L, Károlyi K, Méhes G. Platelet derived growth factor receptor-beta (PDGFRβ) expression is limited to activated stromal cells in the bone marrow and shows a strong correlation with the grade of myelofibrosis. Virchows Arch 2013; 463:57-65. [PMID: 23748876 DOI: 10.1007/s00428-013-1434-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 04/10/2013] [Accepted: 05/24/2013] [Indexed: 12/15/2022]
Abstract
Platelet derived growth factor receptor (PDGFR) is a membrane tyrosine-kinase receptor required for fibroblast activation in stromal proliferations. In order to assess the role of PDGFR in myelofibrosis (MF) we determined in 60 bone marrow biopsies the occurrence and distribution of its α and β subunits in normal and fibrotic bone marrow stroma using immunohistochemistry, and compared this with the grade of MF determined by Gömöri's silver impregnation. PDGF receptor subunits were found to be differentially expressed in the marrow parenchyma. PDGFRα expression identified megakaryocytes, endosteal and endothelial cells while PDGFRβ was virtually absent from inter-trabecular spaces in normal marrow. Activated fibroblasts characteristic for MF intensely expressed PDGFRβ but only a moderate increase in PDGFRα expression was seen. Semi-quantitative PDGFRβ immunoreactivity scores correlated well with the grade of MF in the vast majority of the MF cases (Spearman r= 0.83). Altogether, 21/60 (35.0%) cases showed a relative increase of PDGFRβ expression, compared to the MF grade, suggesting that increased stromal PDGFRβ expression occurs early and precedes reticulin and collagen fiber production during fibroblast activation. In conclusion, bone marrow PDGFRβ expression closely correlates with the grade of MF. Increased immunoreactivity for PDGFRβ occurs already in the prefibrotic stage of the disease and might allow a functional classification of the bone marrow stromal reaction.
Collapse
Affiliation(s)
- Judit Bedekovics
- Department of Pathology, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary
| | | | | | | | | |
Collapse
|
11
|
Kono SA, Heasley LE, Doebele RC, Camidge DR. Adding to the mix: fibroblast growth factor and platelet-derived growth factor receptor pathways as targets in non-small cell lung cancer. Curr Cancer Drug Targets 2012; 12:107-23. [PMID: 22165970 DOI: 10.2174/156800912799095144] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 09/27/2011] [Accepted: 10/11/2011] [Indexed: 01/20/2023]
Abstract
The treatment of advanced non � small cell lung cancer (NSCLC) increasingly involves the use of molecularly targeted therapy with activity against either the tumor directly, or indirectly, through activity against host-derived mechanisms of tumor support such as angiogenesis. The most well studied signaling pathway associated with angiogenesis is the vascular endothelial growth factor (VEGF) pathway, and the only antiangiogenic agent currently approved for the treatment of NSCLC is bevacizumab, an antibody targeted against VEGF. More recently, preclinical data supporting the role of fibroblast growth factor receptor (FGFR) and platelet-derived growth factor receptor (PDGFR) signaling in angiogenesis have been reported. The platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) pathways may also stimulate tumor growth directly through activation of downstream mitogenic signaling cascades. In addition, 1 or both of these pathways have been associated with resistance to agents targeting the epidermal growth factor receptor (EGFR) and VEGF. A number of agents that target FGF and/or PDGF signaling are now in development for the treatment of NSCLC. This review will summarize the potential molecular roles of PDGFR and FGFR in tumor growth and angiogenesis, as well as discuss the current clinical status of PDGFR and FGFR inhibitors in clinical development.
Collapse
Affiliation(s)
- S A Kono
- Aerodigestive and Thoracic Tumor Program, Winship Cancer Institute, Emory University, Room C3005, 1365 Clifton Road NE, Atlanta, GA 30322, USA.
| | | | | | | |
Collapse
|
12
|
Verstraelen S, Nelissen I, Hooyberghs J, Witters H, Schoeters G, Van Cauwenberge P, Van Den Heuvel R. Gene profiles of THP-1 macrophages after in vitro exposure to respiratory (non-)sensitizing chemicals: Identification of discriminating genetic markers and pathway analysis. Toxicol In Vitro 2009; 23:1151-62. [DOI: 10.1016/j.tiv.2009.06.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 05/04/2009] [Accepted: 06/09/2009] [Indexed: 11/30/2022]
|
13
|
Kazlauskas A. Platelet-Derived Growth Factor. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
14
|
Choudhury GG, Mahimainathan L, Das F, Venkatesan B, Ghosh-Choudhury N. c-Src couples PI 3 kinase/Akt and MAPK signaling to PDGF-induced DNA synthesis in mesangial cells. Cell Signal 2007; 18:1854-64. [PMID: 16530387 DOI: 10.1016/j.cellsig.2006.02.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2006] [Revised: 02/02/2006] [Accepted: 02/03/2006] [Indexed: 10/24/2022]
Abstract
Platelet-derived growth factor BB (PDGF) and PDGF receptor-beta (PDGFR) play critical roles in mesangial cell proliferation during embryonic development and in mesangioproliferative glomerulonephritis. We have shown previously that phosphatidylinositol (PI) 3 kinase/Akt and Erk1/2 mitogen-activated protein kinase (MAPK) contribute to PDGF-dependent proliferation of mesangial cells, but the mechanism by which these two enzyme cascades are activated by PDGFR signaling is not precisely known. We examined the role of c-Src tyrosine kinase in this process. PDGF increased phosphorylation of c-Src in a time-dependent manner indicating its activation. A pharmacologic inhibitor of c-Src, PP1, blocked PDGF-induced DNA synthesis with concomitant inhibition of c-Src phosphorylation. Immune-complex kinase assays of c-Src and PDGFR demonstrated inhibition of c-Src tyrosine kinase activity by PP1, without an effect on PDGFR tyrosine phosphorylation. Both PP1 and expression of dominant negative c-Src inhibited PDGF-induced PI 3 kinase, resulting in attenuation of Akt kinase activity. Expression of constitutively active c-Src increased Akt activity to the same extent as with PDGF. Constitutively active c-Src augmented PDGF-induced Akt activity, thus contributing to Akt signaling. Inhibition of c-Src tyrosine kinase blocked PDGF-stimulated MAPK activity and resulted in attenuation of c-fos gene transcription with concomitant prevention of Elk-1 transactivation. Furthermore, inhibition of c-Src increased p27(Kip1) cyclin kinase inhibitor, and attenuated PDGF-induced pRb phosphorylation and CDK2 activity. These data provide the first evidence in mesangial cells that PDGF-activated c-Src tyrosine kinase relays signals to PI 3 kinase/Akt and MAPK. Furthermore our results demonstrate that c-Src integrates signals into the nucleus to activate CDK2, which is required for DNA synthesis.
Collapse
Affiliation(s)
- Goutam Ghosh Choudhury
- Department of Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| | | | | | | | | |
Collapse
|
15
|
Mitchell D, O'Meara SJ, Gaffney A, Crean JKG, Kinsella BT, Godson C. The Lipoxin A4 receptor is coupled to SHP-2 activation: implications for regulation of receptor tyrosine kinases. J Biol Chem 2007; 282:15606-18. [PMID: 17403678 DOI: 10.1074/jbc.m611004200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mesangial cell proliferation is pivotal to the pathology of glomerular injury in inflammation. We have previously reported that lipoxins, endogenously produced eicosanoids with anti-inflammatory and pro-resolution bioactions, can inhibit mesangial cell proliferation in response to several agents. This process is associated with elaborate receptor cross-talk involving modification receptor tyrosine kinase phosphorylation (McMahon, B., Mitchell, D., Shattock, R., Martin, F., Brady, H. R., and Godson, C. (2002) FASEB J. 16, 1817-1819). Here we demonstrate that the lipoxin A(4) (LXA(4)) receptor is coupled to activation and recruitment of the SHP-2 (SH2 domain-containing tyrosine phosphatase-2) within a lipid raft microdomain. Using site-directed mutagenesis of the cytosolic domain of the platelet-derived growth factor receptor beta (PDGFRbeta), we report that mutation of the sites for phosphatidylinositol 3-kinase (Tyr(740) and Tyr(751)) and SHP-2 (Tyr(763) and Tyr(1009)) recruitment specifically inhibit the effect of LXA(4) on the PDGFRbeta signaling; furthermore inhibition of SHP-2 expression with short interfering RNA constructs blocked the effect of LXA(4) on PDGFRbeta phosphorylation. We demonstrate that association of the PDGFRbeta with lipid raft microdomains renders it susceptible to LXA(4)-mediated dephosphorylation by possible reactivation of oxidatively inactivated SHP-2. These data further elaborate on the potential mechanisms underlying the anti-inflammatory, proresolution, and anti-fibrotic bioactions of lipoxins.
Collapse
MESH Headings
- Cell Line
- Cell Proliferation
- Glomerular Mesangium/enzymology
- Glomerular Mesangium/injuries
- Humans
- Inflammation/enzymology
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Membrane Microdomains/enzymology
- Membrane Microdomains/genetics
- Mutagenesis, Site-Directed
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation
- Protein Phosphatase 2
- Protein Processing, Post-Translational/genetics
- Protein Structure, Tertiary/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 11
- Protein Tyrosine Phosphatases/genetics
- Protein Tyrosine Phosphatases/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptors, Formyl Peptide/genetics
- Receptors, Formyl Peptide/metabolism
- Receptors, Lipoxin/genetics
- Receptors, Lipoxin/metabolism
- SH2 Domain-Containing Protein Tyrosine Phosphatases
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Derick Mitchell
- School of Medicine and Medical Science, Diabetes Research Centre, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|
16
|
Rolny C, Nilsson I, Magnusson P, Armulik A, Jakobsson L, Wentzel P, Lindblom P, Norlin J, Betsholtz C, Heuchel R, Welsh M, Claesson-Welsh L. Platelet-derived growth factor receptor-beta promotes early endothelial cell differentiation. Blood 2006; 108:1877-86. [PMID: 16690964 DOI: 10.1182/blood-2006-04-014894] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Platelet-derived growth factor BB (PDGF-BB) has been assigned a critical role in vascular stability by promoting the recruitment of PDGF receptor-beta-expressing perivascular cells. Here we present data indicating that early hematopoietic/endothelial (hemangio) precursors express PDGFR-beta based on coexpression with CD31, vascular endothelial growth factor receptor-2, and CD41 in 2 models: mouse yolk sac (embryonic day 8 [E8]) and differentiating mouse embryonic stem cells (embryoid bodies). Expression of PDGFR-beta on hemangioprecursor cells in the embryoid bodies gradually disappeared, and, at E14, expression appeared on perivascular cells. Activation of the PDGFR-beta on the hemangioprecursors accelerated the differentiation of endothelial cells, whereas differentiation of the hematopoietic lineage was suppressed. In E9.5 yolk sacs derived from recombinant mice expressing kinase-active PDGFR-beta with an aspartic acid to asparagine (D894N) replacement in the kinase activating loop and from mice with ubiquitous expression of PDGF-BB driven by the Rosa26 locus, the number of CD41-expressing early hematopoietic cells decreased by 36% and 34%, respectively, compared with staged wild-type littermates. Moreover, enhanced vascular remodeling was evident in the Rosa26-PDGF-BB yolk sacs. We conclude that PDGFR-beta is expressed on early hemangioprecursor cells, regulating vascular/hematopoietic development.
Collapse
Affiliation(s)
- Charlotte Rolny
- Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv. 20, 751 85 Uppsala, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Rahmani Z. APRO4 negatively regulates Src tyrosine kinase activity in PC12 cells. J Cell Sci 2006; 119:646-58. [PMID: 16434477 DOI: 10.1242/jcs.02778] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Src nonreceptor tyrosine kinase plays an important role in multiple signalling pathways that regulate several cellular functions including proliferation, differentiation and transformation. The activity of Src is tightly regulated in vivo and can be modulated by interactions of its SH2 and SH3 domains with high-affinity ligands. APRO4 (anti-proliferative 4) belongs to a new antiproliferative gene family involved in the negative control of the cell cycle. This report shows that APRO4 associates with Src via its C-terminal proline-rich domain, and downregulates Src kinase activity. Moreover, overexpression of APRO4 leads to inhibition of neurite outgrowth and Ras/MAP kinase signalling in PC12 cells. Furthermore, the kinetics of endogenous Src inactivation correlates with an increase in endogenous APRO4 co-immunoprecipitation in FGF-stimulated PC12 cells. Finally, downregulation of endogenous APRO4 by expression of antisense RNA induces the activation of Src and spontaneous formation of neurites in PC12 cells. Therefore, by controlling the basal threshold of Src activity, APRO4 constitutes an important negative regulatory mechanism for Src-mediated signalling.
Collapse
Affiliation(s)
- Zohra Rahmani
- INSERM U584, Faculté de Médecine Necker-Enfants Malades, 156 Rue de Vaugirard, 75730 Paris CEDEX 15, France.
| |
Collapse
|
18
|
Lee JW, Kim JE, Park EJ, Kim JH, Lee CH, Lee SR, Kwon J. Two conserved cysteine residues are critical for the enzymic function of the human platelet-derived growth factor receptor-beta: evidence for different roles of Cys-822 and Cys-940 in the kinase activity. Biochem J 2005; 382:631-9. [PMID: 15175016 PMCID: PMC1133820 DOI: 10.1042/bj20040624] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2004] [Accepted: 06/03/2004] [Indexed: 02/07/2023]
Abstract
The platelet-derived growth factor receptor-beta (PDGFR-beta) has a number of conserved cysteine residues on its cytoplasmic domain. We have examined whether the cysteine residues play a role in the enzymic function of PDGFR-beta. We found that N-ethylmaleimide, which selectively alkylates free thiol groups of cysteine residues, completely inhibited the kinase activity of PDGFR-beta. We then identified, through site-directed mutagenesis, two conserved cysteine residues critical for the enzymic function of PDGFR-beta. Cys to Ser mutations for either Cys-822, positioned in the catalytic loop, or Cys-940, located in the C-terminal kinase subdomain, significantly reduced the activities of autophosphorylation and phosphorylation towards exogenous substrates. The non-reducing gel analysis indicated that neither of these cysteine residues contributes to the kinase activity by disulphide-bond formation. In addition, the individual mutation of Cys-822 and Cys-940 had no effect on protein stability or the binding of substrates or ATP, implying that these cysteine residues are involved in enzyme catalysis. Finally, proteolytic cleavage assays showed that the mutation of Cys-940, but not Cys-822, induced a protein conformational change. Taken together, these results suggest that Cys-940 contributes to the catalytic activity of PDGFR-beta by playing a structural role, whereas Cys-822 contributes through a different mechanism.
Collapse
Affiliation(s)
- Joon-Won Lee
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Seoul 120-750, South Korea
| | - Jee-Eun Kim
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Seoul 120-750, South Korea
| | - Eun-Jung Park
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Seoul 120-750, South Korea
| | - Jin-Hyun Kim
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Seoul 120-750, South Korea
| | - Chang-Hun Lee
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Seoul 120-750, South Korea
| | - Seung-Rock Lee
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Seoul 120-750, South Korea
| | - Jongbum Kwon
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Seoul 120-750, South Korea
- To whom correspondence should be addressed (email )
| |
Collapse
|
19
|
Abstract
Since its discovery over three decades ago, platelet-derived growth factor (PDGF) has been a model system for learning how growth factors regulate biological processes. For the first several decades investigators used cells grown in tissue culture. More recently, PDGF signaling has also been investigated in mice. This review outlines the advances in these two systems, and highlights some of the directions for future investigation.
Collapse
Affiliation(s)
- Michelle Tallquist
- Deptartment of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | | |
Collapse
|
20
|
James MF, Beauchamp RL, Manchanda N, Kazlauskas A, Ramesh V. A NHERF binding site links the betaPDGFR to the cytoskeleton and regulates cell spreading and migration. J Cell Sci 2004; 117:2951-61. [PMID: 15161943 DOI: 10.1242/jcs.01156] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Na(+)/H(+) exchanger regulatory factor, NHERF, is a multifunctional adapter protein involved in a wide range of physiological activities. NHERF associates with merlin and the ezrin/radixin/moesin (MERM) family of membrane-actin cytoskeletal linker proteins through its C-terminus and is capable of interacting via its PDZ1 domain to the betaPDGF receptor (betaPDGFR). Thus, NHERF, potentially links the betaPDGFR to the actin cytoskeleton through its interaction with MERM proteins. In the present study, we have examined whether abolishing the interaction of betaPDGFR with NHERF results in actin cytoskeletal rearrangements. We have stably expressed a wild-type betaPDGFR, a mutant betaPDGFR (L1106A) that is incapable of interacting with NHERF, as well as a kinase defective mutant receptor (K634R), in PDGFR-deficient mouse embryonic fibroblasts. Our observations indicate that cells expressing betaPDGFR (L1106A) were impaired in their ability to spread and migrate on fibronectin compared with wild-type and K634R cells. L1106A mutant cells also revealed an increased number of focal adhesions, a condensed F-actin ring at the cell periphery and a decrease in total focal adhesion kinase (FAK) tyrosine phosphorylation. Further, we show that NHERF and MERM proteins could act as intermediary bridging proteins between betaPDGFR and FAK. Thus, the interaction of betaPDGFR with NHERF may provide an essential link between the cell membrane and the cortical actin cytoskeleton independent of receptor activity.
Collapse
Affiliation(s)
- Marianne F James
- Molecular Neurogenetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, MA 02129, USA
| | | | | | | | | |
Collapse
|
21
|
Agazie YM, Hayman MJ. Molecular mechanism for a role of SHP2 in epidermal growth factor receptor signaling. Mol Cell Biol 2003; 23:7875-86. [PMID: 14560030 PMCID: PMC207628 DOI: 10.1128/mcb.23.21.7875-7886.2003] [Citation(s) in RCA: 226] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2003] [Revised: 06/16/2003] [Accepted: 07/29/2003] [Indexed: 11/20/2022] Open
Abstract
The Src homology 2-containing phosphotyrosine phosphatase (SHP2) is primarily a positive effector of receptor tyrosine kinase signaling. However, the molecular mechanism by which SHP2 effects its biological function is unknown. In this report, we provide evidence that defines the molecular mechanism and site of action of SHP2 in the epidermal growth factor-induced mitogenic pathway. We demonstrate that SHP2 acts upstream of Ras and functions by increasing the half-life of activated Ras (GTP-Ras) in the cell by interfering with the process of Ras inactivation catalyzed by Ras GTPase-activating protein (RasGAP). It does so by inhibition of tyrosine phosphorylation-dependent translocation of RasGAP to the plasma membrane, to its substrate (GTP-Ras) microdomain. Inhibition is achieved through the dephosphorylation of RasGAP binding sites at the level of the plasma membrane. We have identified Tyr992 of the epidermal growth factor receptor (EGFR) to be one such site, since its mutation to Phe renders the EGFR refractory to the effect of dominant-negative SHP2. To our knowledge, this is the first report to outline the site and molecular mechanism of action of SHP2 in EGFR signaling, which may also serve as a model to describe its role in other receptor tyrosine kinase signaling pathways.
Collapse
Affiliation(s)
- Yehenew M Agazie
- Department of Molecular Genetics and Microbiology, Health Sciences Center, Stony Brook University, Stony Brook, New York 11794-5222, USA
| | | |
Collapse
|
22
|
Chiarugi P, Cirri P, Taddei ML, Giannoni E, Fiaschi T, Buricchi F, Camici G, Raugei G, Ramponi G. Insight into the role of low molecular weight phosphotyrosine phosphatase (LMW-PTP) on platelet-derived growth factor receptor (PDGF-r) signaling. LMW-PTP controls PDGF-r kinase activity through TYR-857 dephosphorylation. J Biol Chem 2002; 277:37331-8. [PMID: 12149261 DOI: 10.1074/jbc.m205203200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Low molecular weight phosphotyrosine phosphatase (LMW-PTP) is an enzyme involved in platelet-derived growth factor-induced mitogenesis and cytoskeleton rearrangement. Our previous results demonstrated that LMW-PTP is able to bind and dephosphorylate activated platelet-derived growth factor receptor (PDGF-r), thus inhibiting cell proliferation. Here we revisit the role of LMW-PTP on activated PDGF-r dephosphorylation. We demonstrate that LMW-PTP preferentially acts on cell surface PDGF-r, excluding the internalized activated receptor pool. Many phosphotyrosine phosphatases act by site-selective dephosphorylation on several sites of PDGF-r, but until now, there has been no evidence of a direct involvement of a specific phosphotyrosine phosphatase in the dephosphorylation of the 857 kinase domain activation tyrosine. Here we report that LMW-PTP affects the kinase activity of the receptor through the binding and dephosphorylation of Tyr-857 and influences many of the signal outputs from the receptor. In particular, we demonstrate a down-regulation of phosphatidylinositol 3-kinase, Src homology phosphatase-2, and phospholipase C-gamma1 binding but not of MAPK activation. In addition, we report a slight action of LMW-PTP on Tyr-716, which directs MAPK activation through Grb2 binding. On the basis of these results, we propose a key role for LMW-PTP in PDGF-r down-regulation through the dephosphorylation of the activation loop Tyr-857, thus determining a general negative regulation of all downstream signals, with the exception of those elicited by internalized receptors.
Collapse
Affiliation(s)
- Paola Chiarugi
- Department of Biochemical Sciences, University of Florence, 50134 Florence, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Saucier C, Papavasiliou V, Palazzo A, Naujokas MA, Kremer R, Park M. Use of signal specific receptor tyrosine kinase oncoproteins reveals that pathways downstream from Grb2 or Shc are sufficient for cell transformation and metastasis. Oncogene 2002; 21:1800-11. [PMID: 11896612 DOI: 10.1038/sj.onc.1205261] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2001] [Revised: 11/02/2001] [Accepted: 12/12/2001] [Indexed: 11/08/2022]
Abstract
Many human cancers have been associated with the deregulation of receptor tyrosine kinases (RTK). However, the individual contribution of receptor-associated signaling proteins in cellular transformation and metastasis is poorly understood. To examine the role of RTK activated signal transduction pathways to processes involved in cell transformation, we have exploited the oncogenic derivative of the Met RTK (Tpr-Met). Unlike other RTKs, twin tyrosine residues in the carboxy-terminal tail of the Met oncoprotein and receptor are required for all biological and transforming activities, and a mutant lacking these tyrosines is catalytically active but non transforming. Using this mutant we have inserted oligonucleotide cassettes, each encoding a binding site for a specific signaling protein derived from other RTKs. We have generated variant forms of the Tpr-Met oncoprotein with the ability to bind individually to the p85 subunit of PI3'K, PLCgamma, or to the Grb2 or Shc adaptor proteins. Variants that recruit the Shc or Grb2 adaptor proteins generated foci of morphologically transformed fibroblast cells and induced anchorage-independent growth, scattering of epithelial cells and experimental metastasis. In contrast, variants that bind and activate PI3'K or PLCgamma failed to generate readily detectable foci. Although cell lines expressing the PI3'K variant grew in soft-agar, these cells were non metastatic. Using this unique RTK oncoprotein model, we have established that Grb2 or Shc dependent signaling pathways are sufficient for cell transformation and metastatic spread.
Collapse
Affiliation(s)
- Caroline Saucier
- Molecular Oncology Group, McGill University Hospital Center, Montreal, Quebec, Canada H3A 1A1
| | | | | | | | | | | |
Collapse
|
24
|
Klinghoffer RA, Hamilton TG, Hoch R, Soriano P. An allelic series at the PDGFalphaR locus indicates unequal contributions of distinct signaling pathways during development. Dev Cell 2002; 2:103-13. [PMID: 11782318 DOI: 10.1016/s1534-5807(01)00103-4] [Citation(s) in RCA: 144] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A central issue in signal transduction is the physiological contribution of different growth factor-initiated signaling pathways. We have generated knockin mice harboring mutations in the PDGFalpha receptor (PDGFalphaR) that selectively eliminate its capacity to activate PI3 kinase (alpha(PI3K)) or Src family kinases (alpha(Src)). The alpha(PI3K) mutation leads to neonatal lethality due to impaired signaling in many cell types, but the alpha(Src) mutation only affects oligodendrocyte development. A third knockin line containing mutations that eliminate multiple docking sites does not increase the severity of the alpha(PI3K) mutation. However, embryos with mutations in the PI3K binding sites of both PDGFRs (alpha and beta) recapitulate the PDGFalphaR null phenotype. Our results indicate that PI3K has a predominant role in PDGFalphaR signaling in vivo and that RTK-activated signaling pathways execute both specific and overlapping functions during mammalian development.
Collapse
Affiliation(s)
- Richard A Klinghoffer
- Program in Developmental Biology, Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | | | | | | |
Collapse
|
25
|
Balciunaite E, Kazlauskas A. Early phosphoinositide 3-kinase activity is required for late activation of protein kinase Cepsilon in platelet-derived-growth-factor-stimulated cells: evidence for signalling across a large temporal gap. Biochem J 2001; 358:281-5. [PMID: 11513725 PMCID: PMC1222059 DOI: 10.1042/0264-6021:3580281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
At least two signalling systems have the potential to contribute to the activation of protein kinase C (PKC) family members such as PKCepsilon. One of these is phosphoinositide 3-kinase (PI 3-kinase), whose lipid products activate PKCepsilon in vitro and in living cells. The recent observation that there are multiple waves of PI 3-kinase and PKCepsilon activity within the G(0)-to-S phase interval provides a new opportunity to investigate the relationship between these two signalling enzymes in vivo. We have assessed the relative importance of the early and late waves of PI 3-kinase activity for the corresponding waves of PKCepsilon activity. Blocking the first phase of PI 3-kinase activity inhibited both early and late activation of PKCepsilon. In contrast, the second wave of PI 3-kinase activity was dispensable for late activation of PKCepsilon. These findings suggested that early PI 3-kinase activation induced a stable change in PKCepsilon, which predisposed it to subsequent activation by lipid cofactors. Indeed, partial proteolysis of PKCepsilon indicated that early activation of PI 3-kinase led to a conformation change in PKCepsilon that persisted as the activity of PKCepsilon cycled. We propose a two-step hypothesis for the activation of PKCepsilon in vivo. One step is stable and depends on PI 3-kinase, whereas the other is transient and may depend on the availability of lipid cofactors. Finally, these studies reveal that PI 3-kinase and PKCepsilon are capable of communicating over a relatively long time interval and begin to elucidate the mechanism.
Collapse
Affiliation(s)
- E Balciunaite
- Institute of Biotechnology, Graiciuno 8, 2028 Vilnius, Lithuania
| | | |
Collapse
|
26
|
Nickas ME, Bernard A, Kazlauskas A. The requirement of tyrosines 579 and 581 for maximal ligand-dependent activation of the betaPDGFR is influenced by noncytoplasmic regions of the receptor. Exp Cell Res 2001; 265:80-9. [PMID: 11281646 DOI: 10.1006/excr.2001.5169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mutating tyrosines 579 and 581 of the beta platelet-derived growth factor receptor (betaPDGFR) tyrosine kinase to phenylalanines (the F2 mutation) impair activation of the receptor in response to ligand, but mutation of the analogous tyrosines in the alphaPDGFR has no effect on ligand-dependent receptor activation. We have found that the F2 mutation has only a modest effect on ligand-dependent activation of a chimeric PDGFR composed of the extracellular and transmembrane domains of the alphaPDGFR and the cytoplasmic domain of the betaPDGFR by three measures: (1) the ability to phosphorylate endogenous and exogenous protein substrates in vitro, (2) phosphorylation of tyrosine 857, and (3) binding of the effector proteins PLCgamma, RasGAP, and SHP-2. Conversely, the F2 mutation substantially impairs ligand-dependent activation of chimeric PDGFRs that consist of either the extracellular domain alone or the extracellular and transmembrane domains of the betaPDGFR and all remaining sequence from the alphaPDGFR by two measures: (1) phosphorylation of endogenous protein substrates in vitro and (2) binding of PLCgamma and SHP-2. Our results indicate that the requirement of tyrosines 579 and 581 for maximal activation of the betaPDGFR in response to ligand is primarily determined by noncytoplasmic regions of the receptor.
Collapse
Affiliation(s)
- M E Nickas
- The Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | |
Collapse
|
27
|
Klinghoffer RA, Mueting-Nelsen PF, Faerman A, Shani M, Soriano P. The two PDGF receptors maintain conserved signaling in vivo despite divergent embryological functions. Mol Cell 2001; 7:343-54. [PMID: 11239463 DOI: 10.1016/s1097-2765(01)00182-4] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Gene targeting studies have indicated that the two receptors for PDGF, alpha and beta, direct unique functions during development. Distinct ligand affinities, patterns of gene expression, and/or mechanisms of signal relay may account for functional specificity of the two PDGF receptor isoforms. To distinguish between these factors, we have created two complementary lines of knockin mice in which the intracellular signaling domains of one PDGFR have been removed and replaced by those of the other PDGFR. While both lines demonstrated substantial rescue of normal development, substitution of the PDGFbetaR signaling domains with those of the PDGFalphaR resulted in varying degrees of vascular disease. This observation provides a framework for discussing the evolution of receptor tyrosine kinase functional specificity.
Collapse
Affiliation(s)
- R A Klinghoffer
- Program in Developmental Biology and Division, Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | |
Collapse
|
28
|
Lai CC, Henningson C, DiMaio D. Bovine papillomavirus E5 protein induces the formation of signal transduction complexes containing dimeric activated platelet-derived growth factor beta receptor and associated signaling proteins. J Biol Chem 2000; 275:9832-40. [PMID: 10734138 DOI: 10.1074/jbc.275.13.9832] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The bovine papillomavirus E5 protein binds to the cellular platelet-derived growth factor (PDGF) beta receptor, resulting in constitutive activation of the receptor and cell growth transformation. By subjecting extracts from E5-transformed or PDGF-treated cells to velocity sedimentation in sucrose gradients, activated PDGF beta receptor complexes were separated from monomeric, inactive receptor. Rapidly sedimenting activated complexes contained oligomeric (apparently dimeric), tyrosine-phosphorylated PDGF beta receptor, the E5 protein, and associated cellular signaling proteins including the p85 subunit of phosphoinositol 3'-kinase, phospholipase Cgamma, and Ras-GTPase activating protein. These signaling proteins made the major contribution to the increased sedimentation rate of the activated receptor complexes. Pairwise analysis of components of these complexes indicated that multiple signaling proteins and the E5 protein were simultaneously present in the activated complexes. Our results also showed that the E5 protein and PDGF activated only a small fraction of the total PDGF beta receptor, that not all receptor molecules associated with the E5 protein were tyrosine-phosphorylated, and that signaling proteins could bind to hemiphosphorylated receptor dimers. On the basis of these results, we propose a model for the assembly of multiprotein, activated PDGF beta receptor complexes in response to the E5 protein.
Collapse
Affiliation(s)
- C C Lai
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | | | |
Collapse
|
29
|
Platelet-derived growth factor-dependent association of the GTPase-activating protein of Ras and Src. Biochem J 2000. [PMID: 10567236 DOI: 10.1042/bj3440519] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Here we report that the platelet-derived growth factor beta receptor (betaPDGFR) is not the only tyrosine kinase able to associate with the GTPase-activating protein of Ras (RasGAP). The interaction of non-betaPDGFR kinase(s) with RasGAP was dependent on stimulation with platelet-derived growth factor (PDGF) and seemed to require tyrosine phosphorylation of RasGAP. Because the tyrosine phosphorylation site of RasGAP is in a sequence context that is favoured by the Src homology 2 ('SH2') domain of Src family members, we tested the possibility that Src was the kinase that associated with RasGAP. Indeed, Src interacted with phosphorylated RasGAP fusion proteins; immunodepletion of Src markedly decreased the recovery of the RasGAP-associated kinase activity. Thus PDGF-dependent tyrosine phosphorylation of RasGAP results in the formation of a complex between RasGAP and Src. To begin to address the relevance of these observations, we focused on the consequences of the interaction of Src and RasGAP. We found that a receptor mutant that did not activate Src was unable to efficiently mediate the tyrosine phosphorylation of phospholipase Cgamma (PLCgamma). Taken together, these observations support the following hypothesis. When RasGAP is recruited to the betaPDGFR, it is phosphorylated and associates with Src. Once bound to RasGAP, Src is no longer able to promote the phosphorylation of PLCgamma. This hypothesis offers a mechanistic explanation for our previously published findings that the recruitment of RasGAP to the betaPDGFR attenuates the tyrosine phosphorylation of PLCgamma. Finally, these findings suggest a novel way in which RasGAP negatively regulates signal relay by the betaPDGFR.
Collapse
|
30
|
Schlesinger TK, Demali KA, Johnson GL, Kazlauskas A. Platelet-derived growth factor-dependent association of the GTPase-activating protein of Ras and Src. Biochem J 1999; 344 Pt 2:519-26. [PMID: 10567236 PMCID: PMC1220671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Here we report that the platelet-derived growth factor beta receptor (betaPDGFR) is not the only tyrosine kinase able to associate with the GTPase-activating protein of Ras (RasGAP). The interaction of non-betaPDGFR kinase(s) with RasGAP was dependent on stimulation with platelet-derived growth factor (PDGF) and seemed to require tyrosine phosphorylation of RasGAP. Because the tyrosine phosphorylation site of RasGAP is in a sequence context that is favoured by the Src homology 2 ('SH2') domain of Src family members, we tested the possibility that Src was the kinase that associated with RasGAP. Indeed, Src interacted with phosphorylated RasGAP fusion proteins; immunodepletion of Src markedly decreased the recovery of the RasGAP-associated kinase activity. Thus PDGF-dependent tyrosine phosphorylation of RasGAP results in the formation of a complex between RasGAP and Src. To begin to address the relevance of these observations, we focused on the consequences of the interaction of Src and RasGAP. We found that a receptor mutant that did not activate Src was unable to efficiently mediate the tyrosine phosphorylation of phospholipase Cgamma (PLCgamma). Taken together, these observations support the following hypothesis. When RasGAP is recruited to the betaPDGFR, it is phosphorylated and associates with Src. Once bound to RasGAP, Src is no longer able to promote the phosphorylation of PLCgamma. This hypothesis offers a mechanistic explanation for our previously published findings that the recruitment of RasGAP to the betaPDGFR attenuates the tyrosine phosphorylation of PLCgamma. Finally, these findings suggest a novel way in which RasGAP negatively regulates signal relay by the betaPDGFR.
Collapse
Affiliation(s)
- T K Schlesinger
- National Jewish Medical and Research Center, Division of Molecular Signal Transduction, 1400 Jackson Street, Denver, CO 80226, USA
| | | | | | | |
Collapse
|
31
|
Emaduddin M, Ekman S, Rönnstrand L, Heldin CH. Functional co-operation between the subunits in heterodimeric platelet-derived growth factor receptor complexes. Biochem J 1999; 341 ( Pt 3):523-8. [PMID: 10417313 PMCID: PMC1220387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
To determine the importance of the phosphorylation capacity of receptor kinase as well as the ability to serve as docking sites for SH2-domain-containing signal transduction molecules, we established pig aortic endothelial cell lines stably expressing kinase-active platelet-derived growth factor (PDGF) alpha-receptors together with kinase-inactive beta-receptors, and vice versa. After stimulation with PDGF-AB, heterodimeric receptor complexes were formed in which the kinase-inactive receptor was phosphorylated by the kinase-active receptor, although less efficiently than in heterodimers of wild-type receptors. The kinase-active receptor was only minimally phosphorylated. Thus the phosphorylation within the receptor dimer occurred in trans between the components. Analyses of the abilities of heterodimeric receptor complexes of one kinase-active and one kinase-inactive receptor to mediate mitogenicity, chemotaxis and activation of mitogen-activated protein kinase revealed less efficient effects than those of heterodimers of wild-type receptors. Importantly, however, the fact that signalling capacities were retained illustrates a functional co-operation between the two receptor molecules in the dimer, where one receptor provides a functional kinase and the other acts as a substrate and provides docking sites for downstream signalling molecules.
Collapse
Affiliation(s)
- M Emaduddin
- Ludwig Institute for Cancer Research, Box 595, Biomedical Center, S-751 24 Uppsala, Sweden
| | | | | | | |
Collapse
|
32
|
Bioukar EB, Marricco NC, Zuo D, Larose L. Serine phosphorylation of the ligand-activated beta-platelet-derived growth factor receptor by casein kinase I-gamma2 inhibits the receptor's autophosphorylating activity. J Biol Chem 1999; 274:21457-63. [PMID: 10409710 DOI: 10.1074/jbc.274.30.21457] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Platelet-derived growth factor (PDGF) receptors (PDGFRs) are membrane protein-tyrosine kinases that, upon activation, become tyrosine-phosphorylated and associate with numerous SH2 domain-containing molecules involved in mediating signal transduction. In Rat-2 fibroblasts, we have characterized the phosphorylation of the beta-PDGFR following its activation by PDGF. In contrast to tyrosine phosphorylation, which was transient and returned to near basal levels by 30 min, PDGF-stimulated Ser/Thr phosphorylation of the beta-PDGFR was increased by 5 min and remained elevated after 30 min. In vivo, after 5 min of PDGF stimulation, serine phosphorylation of the beta-PDGFR was greatly reduced by CKI-7, a specific inhibitor of casein kinase I (CKI). In vitro, recombinant CKI-gamma2 phosphorylated the ligand-activated beta-PDGFR on serine residues in a CKI-7-sensitive manner and resulted in a marked inhibition of the receptor's autophosphorylating activity. Furthermore, in Rat-2 fibroblasts, expression of hemagglutinin epitope-tagged active CKI-gamma2 resulted in a dramatic decrease in the tyrosine phosphorylation state of the beta-PDGFR in response to PDGF, consistent with receptor inactivation. Our data suggest that upon PDGF stimulation, CKI-gamma2 is activated and/or translocated in proximity to the beta-PDGFR, whereby it phosphorylates the beta-PDGFR on serine residues and negatively regulates its tyrosine kinase activity, leading to receptor inactivation.
Collapse
Affiliation(s)
- E B Bioukar
- Polypeptide Laboratory, Department of Experimental Medicine, McGill University, Montreal, Quebec H3A 2B2, Canada
| | | | | | | |
Collapse
|
33
|
Sachsenmaier C, Sadowski HB, Cooper JA. STAT activation by the PDGF receptor requires juxtamembrane phosphorylation sites but not Src tyrosine kinase activation. Oncogene 1999; 18:3583-92. [PMID: 10380880 DOI: 10.1038/sj.onc.1202694] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Activation of the platelet-derived growth factor (PDGF) receptor tyrosine kinase induces tyrosine phosphorylation of Signal Transducer and Activator of Transcription (STAT) proteins. Since the PDGF receptor also activates the Src tyrosine kinase, it is possible that Src mediates tyrosine phosphorylation of STATs in PDGF-treated cells. Consistent with a role for Src in STAT activation, we found that a PDGF receptor juxtamembrane tyrosine residue required for Src activation is necessary and sufficient for activation of STATs 1 and 3. To test the Src requirement further, we made other mutations in the PDGF receptor juxtamembrane region that increased or decreased Src binding. In epithelial and fibroblast cells, PDGF activated STAT1, 3 and 6 in the absence of detectable binding and activation of Src. In addition, PDGF induced c-myc RNA expression and DNA synthesis even though Src was not detectably activated. The activation of MAP kinase and the induction of c-fos gene expression both correlated with STAT but not Src activation by the receptor. We conclude that juxtamembrane tyrosine phosphorylation is necessary for both Src tyrosine kinase and STAT activation by the betaPDGF receptor, but that both processes are regulated independently by this region.
Collapse
Affiliation(s)
- C Sachsenmaier
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | | | |
Collapse
|
34
|
Klinghoffer RA, Sachsenmaier C, Cooper JA, Soriano P. Src family kinases are required for integrin but not PDGFR signal transduction. EMBO J 1999; 18:2459-71. [PMID: 10228160 PMCID: PMC1171328 DOI: 10.1093/emboj/18.9.2459] [Citation(s) in RCA: 608] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Src family kinases (SFKs) have been implicated as important regulators of ligand-induced cellular responses including proliferation, survival, adhesion and migration. Analysis of SFK function has been impeded by extensive redundancy between family members. We have generated mouse embryos harboring functional null mutations of the ubiquitously expressed SFKs Src, Yes and Fyn. This triple mutation leads to severe developmental defects and lethality by E9.5. To elucidate the molecular mechanisms underlying this phenotype, SYF cells (deficient for Src, Yes and Fyn) were derived and tested for their ability to respond to growth factors or plating on extracellular matrix. Our studies reveal that while Src, Yes and Fyn are largely dispensable for platelet-derived growth factor (PDGF)-induced signaling, they are absolutely required to mediate specific functions regulated by extracellular matrix proteins. Fibronectin-induced tyrosine phosphorylation of focal adhesion proteins, including the focal adhesion kinase FAK, was nearly eliminated in the absence of Src, Yes and Fyn. Furthermore, consistent with previous reports demonstrating the importance of FAK for cell migration, SYF cells displayed reduced motility in vitro. These results demonstrate that SFK activity is essential during embryogenesis and suggest that defects observed in SYF triple mutant embryos may be linked to deficiencies in signaling by extracellular matrix-coupled receptors.
Collapse
Affiliation(s)
- R A Klinghoffer
- Program in Developmental Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North-Mailstop C3-168, PO Box 19204, Seattle, WA 98109-1024, USA
| | | | | | | |
Collapse
|
35
|
Hossain MZ, Jagdale AB, Ao P, Kazlauskas A, Boynton AL. Disruption of gap junctional communication by the platelet-derived growth factor is mediated via multiple signaling pathways. J Biol Chem 1999; 274:10489-96. [PMID: 10187840 DOI: 10.1074/jbc.274.15.10489] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The platelet-derived growth factor (PDGF) mediates its cellular functions via activation of its receptor tyrosine kinase followed by the recruitment and activation of several signaling molecules. These signaling molecules then initiate specific signaling cascades, finally resulting in distinct physiological effects. To delineate the PDGF signaling pathway responsible for the disruption of gap junctional communication (GJC), wild-type PDGF receptor beta (PDGFRbeta) and a series of PDGFRbeta mutants were expressed in T51B rat liver epithelial cells. In cells expressing wild-type PDGFRbeta, PDGF induced disruption of GJC and phosphorylation of a gap junctional protein, connexin-43 (Cx43), which required activation of mitogen-activated protein kinase, although involvement of additional factors was also evident. In the F5 mutant lacking binding sites for phosphatidylinositol 3-kinase, GTPase-activating protein, SHP-2, and phospholipase Cgamma1 (PLCgamma1), PDGF induced mitogen-activated protein kinase, but failed to affect GJC or Cx43, indicating involvement of additional signals presumably initiated by one or more of the mutated binding sites. Examination of the single-site mutants revealed that PDGF effects were not mediated via a single signaling component. This was confirmed by the "add-back" mutants, which showed that restoration of either SHP-2 or PLCgamma1 binding was sufficient to propagate the GJC inhibitory actions of PDGF. Further analysis showed that activation of PLCgamma1 is involved in Cx43 phosphorylation, which surprisingly failed to correlate with GJC blockade. The results of our study demonstrate that PDGF-induced disruption of GJC can be mediated by multiple signaling pathways and requires participation of multiple components.
Collapse
Affiliation(s)
- M Z Hossain
- Molecular Medicine, Northwest Hospital, Seattle, Washington 98125, USA.
| | | | | | | | | |
Collapse
|
36
|
Lai CC, Henningson C, DiMaio D. Bovine papillomavirus E5 protein induces oligomerization and trans-phosphorylation of the platelet-derived growth factor beta receptor. Proc Natl Acad Sci U S A 1998; 95:15241-6. [PMID: 9860953 PMCID: PMC28027 DOI: 10.1073/pnas.95.26.15241] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/1998] [Indexed: 01/09/2023] Open
Abstract
The bovine papillomavirus E5 protein is a 44-aa transmembrane protein that forms a stable complex with the cellular platelet-derived growth factor (PDGF) beta receptor and induces constitutive tyrosine phosphorylation and activation of the receptor, resulting in cell transformation. The E5 protein does not resemble PDGF, but rather activates the receptor in a ligand-independent fashion, thus providing a unique system to examine activation of receptor tyrosine kinases. Here, we used a variety of approaches to explore the mechanism of receptor activation by the E5 protein. Chemical cross-linking experiments revealed that the E5 protein activated only a small fraction of the endogenous PDGF beta receptor in transformed fibroblasts and suggested that this fraction was constitutively dimerized. Coimmunoprecipitation experiments using extracts of cells engineered to coexpress full-length and truncated PDGF beta receptors confirmed that the E5 protein induced oligomerization of the receptor. Furthermore, in cells expressing the E5 protein, a kinase-active receptor was able to trans-phosphorylate a kinase-negative mutant receptor but was unable to catalyze intramolecular autophosphorylation. These results indicated that the E5 protein induced PDGF beta receptor activation by forming a stable complex with the receptor, resulting in receptor dimerization and trans-phosphorylation.
Collapse
Affiliation(s)
- C C Lai
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | | | | |
Collapse
|
37
|
Hess JA, Ji QS, Carpenter G, Exton JH. Analysis of platelet-derived growth factor-induced phospholipase D activation in mouse embryo fibroblasts lacking phospholipase C-gamma1. J Biol Chem 1998; 273:20517-24. [PMID: 9685408 DOI: 10.1074/jbc.273.32.20517] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Platelet-derived growth factor (PDGF) activates phospholipase D (PLD) in mouse embryo fibroblasts (MEFs). In order to investigate a role for phospholipase C-gamma1 (PLC-gamma1), we used targeted disruption of the Plcg1 gene in the mouse to develop Plcg1(+/+) and Plcg1(-/-) cell lines. Plcg1(+/+) MEFs treated with PDGF showed a time- and dose-dependent increase in the production of total inositol phosphates that was substantially reduced in Plcg1(-/-) cells. Plcg1(+/+) cells also showed a PDGF-induced increase in PLD activity that had a similar dose dependence to the PLC response but was down-regulated after 15 min. Phospholipase D activity, however, was markedly reduced in Plcg1(-/-) cells. The PDGF-induced inositol phosphate formation and the PLD activity that remained in the Plcg1(-/-) cells could be attributed to the presence of phospholipase C-gamma2 (PLC-gamma2) in the Plcg1(-/-) cells. The PLC-gamma2 expressed in the Plcg1(-/-) cells was phosphorylated on tyrosine in response to PDGF treatment, and a small but significant fraction of the Plcg1(-/-) cells showed Ca2+ mobilization in response to PDGF, suggesting that the PLC-gamma2 expressed in the Plcg1(-/-) cells was activated in response to PDGF. The inhibition of PDGF-induced phospholipid hydrolysis in Plcg1(-/-) cells was not due to differences in the level of PDGF receptor or in the ability of PDGF to cause autophosphorylation of the receptor. Upon treatment of the Plcg1(-/-) cells with oleoylacetylglycerol and the Ca2+ ionophore ionomycin to mimic the effect of PLC-gamma1, PLD activity was restored. The targeted disruption of Plcg1 did not result in universal changes in the cell signaling pathways of Plcg1(-/-) cells, because the phosphorylation of mitogen-activated protein kinase was similar in Plcg1(+/+) and Plcg1(-/-) cells. Because increased plasma membrane ruffles occurred in both Plcg1(+/+) and Plcg1(-/-) cells following PDGF treatment, it is possible neither PLC nor PLD are necessary for this growth factor response. In summary, these data indicate that PLC-gamma is required for growth factor-induced activation of PLD in MEFs.
Collapse
Affiliation(s)
- J A Hess
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0295, USA
| | | | | | | |
Collapse
|
38
|
Hossain MZ, Ao P, Boynton AL. Platelet-derived growth factor-induced disruption of gap junctional communication and phosphorylation of connexin43 involves protein kinase C and mitogen-activated protein kinase. J Cell Physiol 1998; 176:332-41. [PMID: 9648920 DOI: 10.1002/(sici)1097-4652(199808)176:2<332::aid-jcp11>3.0.co;2-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Previously we showed a rapid and transient inhibition of gap junctional communication (GJC) by platelet-derived growth factor (PDGF) in T51B rat liver epithelial cells expressing wild-type platelet-derived growth factor beta receptors (PDGFrbeta). This action of PDGF correlated with the hyperphosphorylation of the gap junction protein connexin43 (Cx43) and required PDGFrbeta tyrosine kinase activity, suggesting the participation of protein kinases and phosphatases many of which are activated by PDGF treatment. In the present study, two such kinases, namely protein kinase C (PKC) and mitogen-activated protein kinase (MAPK), are investigated for their possible involvement in PDGF-induced closure of junctional channels and Cx43-phosphorylation. Down-regulation of PKC-isoforms by 12-O-tetradecanoylphorbol-13-acetate or pretreatment with the PKC inhibitor calphostin C, completely blocked PDGF action on GJC and Cx43. Activation of MAPK correlated with PDGF-induced Cx43 phosphorylation, and prevention of MAPK activation by PD98059 eliminated the PDGF effects. Interestingly, elimination of GJC recovery by cycloheximide was associated with a sustained activated-MAPK level. Based on these results we postulate that the activation of PKC and MAPK are required in PDGF-mediated Cx43 phosphorylation and junctional closure.
Collapse
Affiliation(s)
- M Z Hossain
- Molecular Medicine, Northwest Hospital, Seattle, Washington 98125, USA.
| | | | | |
Collapse
|
39
|
Baxter RM, Secrist JP, Vaillancourt RR, Kazlauskas A. Full activation of the platelet-derived growth factor beta-receptor kinase involves multiple events. J Biol Chem 1998; 273:17050-5. [PMID: 9642269 DOI: 10.1074/jbc.273.27.17050] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of receptor tyrosine kinases is thought to involve ligand-induced dimerization, which promotes receptor transphosphorylation and thereby increases the receptor's phosphotransferase activity. We used two platelet-derived growth factor beta-receptor (beta-PDGFR) mutants to identify events that are required for full engagement (autophosphorylation and activation of the kinase activity) of the beta-PDGFR kinase. The F79/81 receptor (Tyr to Phe substitution at 579 and 581 in the juxtamembrane domain of the receptor) was capable of only very modest ligand-dependent autophosphorylation and also failed to associate with numerous SH2 domain-containing proteins. Furthermore, stimulation with platelet-derived growth factor (PDGF) did not increase the kinase activity of the F79/81 mutant toward exogenous substrates. However, the F79/81 receptor had basal kinase activity and could be artificially stimulated by incubation with ATP. Because the low kinase activity of the F857 mutant (Tyr to Phe substitution at 857 in the putative activation loop) could not be increased by incubation with ATP, failure to phosphorylate Tyr-857 may be the reason why the F79/81 mutant has low kinase activity. Surprisingly, the F857 mutant underwent efficient PDGF-dependent autophosphorylation. Thus the PDGF-dependent increase in the kinase activity of the receptor is not required for autophosphorylation. We conclude that full activation of the beta-PDGFR kinase requires at least two, apparently distinct events.
Collapse
Affiliation(s)
- R M Baxter
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | |
Collapse
|
40
|
Liu J, Fitzli D, Liu M, Tseu I, Caniggia I, Rotin D, Post M. PDGF-induced glycosaminoglycan synthesis is mediated via phosphatidylinositol 3-kinase. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:L702-13. [PMID: 9612285 DOI: 10.1152/ajplung.1998.274.5.l702] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Platelet-derived growth factor (PDGF)-BB has been shown previously to increase glycosaminoglycan (GAG) synthesis but not DNA synthesis in freshly isolated fetal lung fibroblasts. In the present study, we found that PDGF-BB also enhanced 35SO4 incorporation into the small, soluble proteoglycan biglycan without affecting biglycan's core protein mRNA expression, suggesting that PDGF-BB mainly affects GAG chain elongation and/or sulfation. PDGF-BB-stimulated GAG synthesis was abrogated by tyrphostin 9, a PDGF receptor-associated tyrosine kinase inhibitor, implying that the stimulatory effect is mediated via the PDGF beta-receptor (PDGFR). The intracellular signal transduction pathways that mediate PDGF-BB-stimulated GAG synthesis in fetal lung fibroblasts were investigated. On ligand-induced tyrosine phosphorylation, PDGFR associated with phospholipase C (PLC)-gamma 1, Ras GTPase activating protein (RasGAP), and phosphatidylinositol 3-kinase (PI3K) but not with the Syp-growth factor receptor-bound protein 2-Son of Sevenless complex. Association of PDGFR with PLC-gamma 1 and RasGAP followed by their tyrosine phosphorylation failed, however, to activate PLC-gamma 1, protein kinase C (PKC), and Ras. Neither a PLC-gamma inhibitor, U-73122; a PKC inhibitor, calphostin C; nor a mitogen-activated protein kinase kinase inhibitor, PD-98059, inhibited PDGF-BB-induced GAG synthesis. In contrast, PDGF-BB stimulation triggered PDGFR-associated PI3K activity. Both PDGF-BB-induced PI3K activation and GAG synthesis were abolished by the PI3K inhibitors wortmannin and LY-294002. The results suggest that PI3K is a downstream mediator of PDGF-BB-stimulated GAG synthesis in fetal rat lung fibroblasts.
Collapse
Affiliation(s)
- J Liu
- Department of Pediatrics, Hospital for Sick Children Research Institute, University of Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
41
|
Lin YC, Ho CH, Grinnell F. Decreased PDGF receptor kinase activity in fibroblasts contracting stressed collagen matrices. Exp Cell Res 1998; 240:377-87. [PMID: 9597011 DOI: 10.1006/excr.1998.4013] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fibroblasts cultured in mechanically stressed collagen matrices proliferate, whereas cells in floating collagen matrices become quiescent. Previous research indicated that one factor contributing to cell quiescence in floating matrices was reduced receptor autophosphorylation in response to PDGF stimulation (i.e., PDGF receptor desensitization). To learn more about the mechanism of PDGF receptor desensitization, we analyzed changes in PDGF receptor autophosphorylation and receptor kinase activity after stressed collagen matrices were switched to floating conditions, which results in rapid cell contraction and dissipation of mechanical stress. PDGF receptor desensitization occurred during contraction stimulated by serum but not in the absence of serum, and desensitization was prevented by inhibitors of contraction but not by inhibitors of the contraction-activated cyclic AMP signaling pathway. Receptor desensitization resulted from decreased receptor kinase activity rather than from elevated protein tyrosine phosphatase activity, and only receptors unoccupied at the time of contraction were affected. After contraction, radiolabeled PDGF binding to the cells was decreased, which suggested that receptor desensitization resulted from a contraction-dependent change in receptor availability or affinity.
Collapse
Affiliation(s)
- Y C Lin
- Department of Cell Biology and Neuroscience, University of Texas Southwestern Medical School, Dallas 75235, USA
| | | | | |
Collapse
|
42
|
Habib AA, Högnason T, Ren J, Stefánsson K, Ratan RR. The epidermal growth factor receptor associates with and recruits phosphatidylinositol 3-kinase to the platelet-derived growth factor beta receptor. J Biol Chem 1998; 273:6885-91. [PMID: 9506992 DOI: 10.1074/jbc.273.12.6885] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Receptor tyrosine kinases are classified into subfamilies, which are believed to function independently, with heterodimerization occurring only within the same subfamily. In this study, we present evidence suggesting a direct interaction between the epidermal growth factor (EGF) receptor (EGFR) and the platelet-derived growth factor beta (PDGFbeta) receptor (PDGFbetaR), members of different receptor tyrosine kinase subfamilies. We find that the addition of EGF to COS-7 cells and to human foreskin Hs27 fibroblasts results in a rapid tyrosine phosphorylation of the PDGFbetaR and results in the recruitment of phosphatidylinositol 3-kinase to the PDGFbetaR. In R1hER cells, which overexpress the EGFR, we find ligand-independent tyrosine phosphorylation of the PDGFbetaR and the constitutive binding of a substantial amount of PI-3 kinase activity to it, mimicking the effect of ligand in untransfected cells. In support of the possibility that this may be a direct interaction, we show that the two receptors can be coimmunoprecipitated from untransfected Hs27 fibroblasts and from COS-7 cells. This association can be reconstituted by introducing the two receptors into 293 EBNA cells. The EGFR/PDGFbetaR association is ligand-independent in all cell lines tested. We also demonstrate that the fraction of PDGFbetaR bound to the EGFR in R1hER cells undergoes an EGF-induced mobility shift on Western blots indicative of phosphorylation. Our findings indicate that direct interactions between receptor tyrosine kinases classified under different subfamilies may be more widespread than previously believed.
Collapse
Affiliation(s)
- A A Habib
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | |
Collapse
|
43
|
Hossain MZ, Ao P, Boynton AL. Rapid disruption of gap junctional communication and phosphorylation of connexin43 by platelet-derived growth factor in T51B rat liver epithelial cells expressing platelet-derived growth factor receptor. J Cell Physiol 1998; 174:66-77. [PMID: 9397157 DOI: 10.1002/(sici)1097-4652(199801)174:1<66::aid-jcp8>3.0.co;2-e] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Gap junctional communication (GJC) between contacting cells has been postulated to be involved in the regulation of cell proliferation. This suggestion stems from numerous studies showing modulation of GJC by agents that influence cellular proliferation. Platelet-derived growth factor (PDGF), a strong mitogen, inhibits GJC in many cell types. To understand the molecular nature of the signal transduction pathway responsible for the GJC blockade, T51B rat liver epithelial cells, which lack endogenous PDGF receptor (PDGFr), were infected with a retrovirus containing either wild-type full-length cDNA of human PDGFr beta (Kin+) or a mutant PDGFr beta lacking receptor tyrosine kinase activity (Kin-). PDGF caused a complete but transient interruption of cell communication in Kin+ cells within 15-20 min of addition. This interruption of GJC was not associated with a gross destabilization of gap junction plaques but with the phosphorylation of connexin43 (Cx43), the only known gap junction protein expressed in these cells. These effects were exhibited in either control T51B cells or in Kin- cells, indicating a requirement of the receptor tyrosine kinase activity. Further examination revealed that the newly phosphorylated Cx43 then undergoes a rapid degradation utilizing the lysosomal pathway resulting in a decreased total Cx43 protein level. The re-establishment of GJC following PDGF treatment was dependent on protein synthesis. This report describes a suitable cell system which is currently being utilized for the characterization of the PDGF signaling pathway responsible for the inhibition of GJC.
Collapse
Affiliation(s)
- M Z Hossain
- Department of Molecular Medicine, Northwest Hospital, Seattle, Washington 98125, USA.
| | | | | |
Collapse
|
44
|
Montmayeur JP, Valius M, Vandenheede J, Kazlauskas A. The platelet-derived growth factor beta receptor triggers multiple cytoplasmic signaling cascades that arrive at the nucleus as distinguishable inputs. J Biol Chem 1997; 272:32670-8. [PMID: 9405485 DOI: 10.1074/jbc.272.51.32670] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Stimulation of the platelet-derived growth factor beta receptor (betaPDGFR) activates enzymes such as phosphatidylinositol 3-kinase (PI3K) and phospholipase Cgamma1 (PLCgamma), which ultimately initiate nuclear responses such as enhanced expression of immediate early genes. In an attempt to compare the signaling cascades initiated by PI3K and PLCgamma, we examined the activation of a panel of immediate early genes by betaPDGFR mutants, which preferentially engage PI3K or PLCgamma. When expressed in A431 cells, the wild type receptor and to a lesser extent the mutant receptor that associates with PLCgamma (Y1021) was able to up-regulate c-fos, junB, and KC mRNA expression. In contrast, the receptor mutant that engages PI3K (Y740/51) poorly stimulated c-fos mRNA expression and did not significantly stimulate expression of either JunB or KC. Receptor mutants that did not associate with either PI3K or PLCgamma were dramatically compromised or unable to increase expression of any of these immediate early genes. The differential ability of the Y1021 and Y740/51 receptors to activate c-fos correlated well with an apparent difference in their ability to engage distinct protein kinase C family members. However there did appear to be a degree of redundancy in the cytoplasmic signaling pathways initiated by PI3K and PLCgamma, since both the Y1021 and Y740/51 receptors were able to activate an AP-1-responsive element. We conclude that recruitment of signal relay enzymes to the betaPDGFR is necessary for PDGF-dependent activation of at least some immediate early genes. In addition, whereas the betaPDGFR activates multiple signaling enzymes capable of activating the same nuclear response (activation of c-fos), these signaling cascades do not appear to converge in the cytoplasm but arrive at the nucleus as distinguishable inputs.
Collapse
Affiliation(s)
- J P Montmayeur
- Schepens Eye Research Institute, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | |
Collapse
|
45
|
Fanger GR, Vaillancourt RR, Heasley LE, Montmayeur JP, Johnson GL, Maue RA. Analysis of mutant platelet-derived growth factor receptors expressed in PC12 cells identifies signals governing sodium channel induction during neuronal differentiation. Mol Cell Biol 1997; 17:89-99. [PMID: 8972189 PMCID: PMC231733 DOI: 10.1128/mcb.17.1.89] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The mechanisms governing neuronal differentiation, including the signals underlying the induction of voltage-dependent sodium (Na+) channel expression by neurotrophic factors, which occurs independent of Ras activity, are not well understood. Therefore, Na+ channel induction was analyzed in sublines of PC12 cells stably expressing platelet-derived growth factor (PDGF) beta receptors with mutations that eliminate activation of specific signalling molecules. Mutations eliminating activation of phosphatidylinositol 3-kinase (PI3K), phospholipase C gamma (PLC gamma), the GTPase-activating protein (GAP), and Syp phosphatase failed to diminish the induction of type II Na+ channel alpha-subunit mRNA and functional Na+ channel expression by PDGF, as determined by RNase protection assays and whole-cell patch clamp recording. However, mutation of juxtamembrane tyrosines that bind members of the Src family of kinases upon receptor activation inhibited the induction of functional Na+ channels while leaving the induction of type II alpha-subunit mRNA intact. Mutation of juxtamembrane tyrosines in combination with mutations eliminating activation of PI3K, PLC gamma, GAP, and Syp abolished the induction of type II alpha-subunit mRNA, suggesting that at least partially redundant signaling mechanisms mediate this induction. The differential effects of the receptor mutations on Na+ channel expression did not reflect global changes in receptor signaling capabilities, as in all of the mutant receptors analyzed, the induction of c-fos and transin mRNAs still occurred. The results reveal an important role for the Src family in the induction of Na+ channel expression and highlight the multiplicity and combinatorial nature of the signaling mechanisms governing neuronal differentiation.
Collapse
Affiliation(s)
- G R Fanger
- Department of Biochemistry, Dartmouth Medical School, Hanover, New Hampshire 03755, USA
| | | | | | | | | | | |
Collapse
|
46
|
Soans C, Holash JA, Pavlova Y, Pasquale EB. Developmental expression and distinctive tyrosine phosphorylation of the Eph-related receptor tyrosine kinase Cek9. J Cell Biol 1996; 135:781-95. [PMID: 8909550 PMCID: PMC2121056 DOI: 10.1083/jcb.135.3.781] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Cek9 is a receptor tyrosine kinase of the Eph subfamily for which only a partial cDNA sequence was known (Sajjadi, F.G., and E.B. Pasquale. 1993. Oncogene. 8:1807-1813). We have obtained the entire cDNA sequence and identified a variant form of Cek9 that lacks a signal peptide. We subsequently examined the spatio-temporal expression and tyrosine phosphorylation of Cek9 in the chicken embryo by using specific antibodies. At embryonic day 2, Cek9 immunoreactivity is concentrated in the eye, the brain, the posterior region of the neural tube, and the most recently formed somites. Later in development, Cek9 expression is widespread but particularly prominent in neural tissues. In the developing visual system, Cek9 is highly concentrated in areas containing retinal ganglion cell axons, suggesting a role in regulating their outgrowth to the optic tectum. Unlike other Eph-related receptors, Cek9 is substantially phosphorylated on tyrosine in many tissues at various developmental stages. Since autophosphorylation of receptor protein-tyrosine kinases typically correlates with increased enzymatic activity, this suggests that Cek9 plays an active role in embryonic signal transduction pathways.
Collapse
Affiliation(s)
- C Soans
- Burnham Institute (formerly La Jolla Cancer Research Foundation), La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
47
|
Klinghoffer RA, Duckworth B, Valius M, Cantley L, Kazlauskas A. Platelet-derived growth factor-dependent activation of phosphatidylinositol 3-kinase is regulated by receptor binding of SH2-domain-containing proteins which influence Ras activity. Mol Cell Biol 1996; 16:5905-14. [PMID: 8816504 PMCID: PMC231592 DOI: 10.1128/mcb.16.10.5905] [Citation(s) in RCA: 105] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Upon binding of platelet-derived growth factor (PDGF), the PDGF beta receptor (PDGFR) undergoes autophosphorylation on distinct tyrosine residues and binds several SH2-domain-containing signal relay enzymes, including phosphatidylinositol 3-kinase (PI3K), phospholipase C gamma (PLC gamma), the GTPase-activating protein of Ras (RasGAP), and the tyrosine phosphatase SHP-2. In this study, we have investigated whether PDGF-dependent PI3K activation is affected by the other proteins that associate with the PDGFR. We constructed and characterized a series of PDGFR mutants which contain binding sites for PI3K as well as one additional protein, either RasGAP, SHP-2, or PLC gamma. While all of the receptors had wild-type levels of PDGF-stimulated tyrosine kinase activity and associated with comparable amounts of PI3K activity, their abilities to trigger accumulation of PI3K products in vivo differed dramatically. The wild-type receptor, as well as receptors that recruited PI3K or PI3K and SHP-2, were all capable of fully activating PI3K. In contrast, receptors that associated with PI3K and RasGAP or PI3K and PLC gamma displayed a greatly reduced ability to stimulate production of PI3K products. When this series of receptors was tested for their ability to activate Ras, we observed a strong positive correlation between Ras activation and PI3K activation. Further investigation of the relationship between Ras and PI3K indicated that Ras was upstream of PI3K. Thus, activation of PI3K requires not only binding of PI3K to the tyrosine-phosphorylated PDGFR but accumulation of GTP-bound Ras as well. Furthermore, PLC gamma and RasGAP negatively modulate PDGF-dependent PI3K activation. Finally, PDGF-stimulated signal relay can be regulated by altering the ratio of SH2-domain-containing enzymes that are recruited to the PDGFR.
Collapse
Affiliation(s)
- R A Klinghoffer
- Division of Basic Sciences, National Jewish Center for Immunology and Respiratory Medicine, Denver, Colorado 80206, USA
| | | | | | | | | |
Collapse
|
48
|
Coats SR, Pledger WJ, Awazu M, Daniel TO. Detergent solubility defines an alternative itinerary for a subpopulation of PDGF beta receptors. J Cell Physiol 1996; 168:412-23. [PMID: 8707877 DOI: 10.1002/(sici)1097-4652(199608)168:2<412::aid-jcp20>3.0.co;2-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Current models of platelet-derived growth factor (PDGF) beta receptor itinerary are based upon the properties of receptors recovered from nonionic detergent-solubilized cellular extracts. Comparing several commonly used cell extraction procedures, we have determined that up to 50% of immunoreactive PDGF beta receptors, reside in a Triton X-100 insoluble pool in a wide distribution of cultured cell lines, including Balb/c-3T3, NIH 3T3, and Swiss fibroblasts, primary murine and human fibroblasts, and primary human glial cells. Many properties of Triton insoluble receptors are distinct from the well-characterized PDGF beta receptors, including 1) delayed arrival of newly synthesized receptors into the Triton insoluble fraction, 2) prolonged half-life in the presence of PDGF, 3) increased abundance with increasing cell density, 4) inaccessibility to modification by extracellular compartment enzymes, 5) cofractionation with cytoskeletal proteins, and 6) a higher basal tyrosine phosphorylation state. PDGF stimulates accumulation of tyrosine phosphorylated PDGF beta receptors in the Triton X-100 insoluble fraction. Cell surface PDGF beta receptors modified by enzymatic desialylation redistribute to the insoluble fraction. These findings distinguish the itinerary of a large subpopulation of PDGF beta receptors from those characterized previously. Receptors in this fraction represent a long-lived tyrosine phosphorylated population that may effect responses for extended periods following ligand activation.
Collapse
Affiliation(s)
- S R Coats
- Department of Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | |
Collapse
|
49
|
Abstract
The activation of the serine/threonine kinase, Raf-1, serves to connect upstream protein tyrosine kinases to downstream signaling events. We previously reported that FcgammaRI stimulation of interferon gamma-differentiated U937 cells (termed U937IF cells) induces a mobility shift in Erk2. Herein, we report that cross-linking of FcgammaRI receptor in U937IF cells induces a marked tyrosine phosphorylation of Raf-1 (10-fold increase). Tyrosine phosphorylation of Raf-1 is induced by FcgammaRI activation and not by PMA (1 microg/ml), N-formyl-Met-Leu-Phe (1 microM), calcium ionophore (1 microM), thrombin (0.05 unit/ml), FcgammaRII, or FcgammaRIII stimulation. The kinetics of Raf-1 tyrosine phosphorylation is rapid, reaching peak levels 1-2 min after FcgammaRI activation, and the tyrosine phosphorylation of Raf-1 precedes the activation of the respiratory burst. FcgammaRI cross-linking induces the tyrosine phosphorylation of Shc; tyrosine-phosphorylated Shc binds to Grb2 forming a Shc-Grb2 complex. The data provide evidence that the FcgammaRI receptor signals via the upstream activation of nonreceptor protein tyrosine kinases, which leads to the subsequent activation of Ras family GTPases and serine/threonine kinases, Raf-1 and mitogen-activated protein kinase.
Collapse
Affiliation(s)
- R K Park
- Neil Bogart Memorial Laboratories, Division of Hematology-Oncology, Childrens Hospital Los Angeles and University of Southern California School of Medicine, Los Angeles, California 90027, USA
| | | | | |
Collapse
|
50
|
Kamikura DM, Naujokas MA, Park M. Identification of tyrosine 489 in the carboxy terminus of the Tpr-Met oncoprotein as a major site of autophosphorylation. Biochemistry 1996; 35:1010-7. [PMID: 8547236 DOI: 10.1021/bi9514065] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The Met receptor tyrosine kinase is the receptor for hepatocyte growth factor/scatter factor. HGF/SF is a multifunctional cytokine that can stimulate proliferation, motility, and morphogenesis in epithelial and endothelial cells. Oncogenic activation of the Met receptor occurs through a genomic rearrangement that generates a hybrid protein in which tpr sequences are directly fused amino terminal to the met receptor kinase domain. The resultant Tpr-Met hybrid protein possesses tyrosine kinase activity, is constitutively phosphorylated on tyrosine residues in vivo, and transforms fibroblasts in culture. We have identified two tyrosine residues within the catalytic domain of the Tpr-Met oncoprotein (Y365, Y366) and Met receptor (Y1234, Y1235) that are phosphorylated and essential for both the catalytic and biological activity of the oncoprotein and receptor. However, a detailed analysis of phosphorylation in these proteins has not been undertaken. In order to determine the sites of tyrosine phosphorylation in the Tpr-Met oncoprotein, in vitro mutagenesis, phosphopeptide mapping, and dephosphorylation protection assays were performed. Here we identify that a single tyrosine (Y489) in the carboxy terminus of the Tpr-Met oncoprotein is highly phosphorylated and is essential for biological activity. In contrast, additional tyrosines (Y482, Y498) located in the carboxy terminus are not phosphorylated at detectable levels and are not essential for the biological activity of the oncoprotein.
Collapse
Affiliation(s)
- D M Kamikura
- Royal Victoria Hospital, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|