51
|
|
52
|
Küppers M, Faust D, Linz B, Dietrich C. Regulation of ERK1/2 activity upon contact inhibition in fibroblasts. Biochem Biophys Res Commun 2011; 406:483-7. [DOI: 10.1016/j.bbrc.2011.02.080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 02/15/2011] [Indexed: 10/18/2022]
|
53
|
Density enhanced phosphatase-1 down-regulates urokinase receptor surface expression in confluent endothelial cells. Blood 2011; 117:4154-61. [PMID: 21304107 DOI: 10.1182/blood-2010-09-307694] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
VEGF(165), the major angiogenic growth factor, is known to activate various steps in proangiogenic endothelial cell behavior, such as endothelial cell migration and invasion, or endothelial cell survival. Thereby, the urokinase-type plasminogen activator (uPA) system has been shown to play an essential role not only by its proteolytic capacities, but also by induction of intracellular signal transduction. Therefore, expression of its cell surface receptor uPAR is thought to be an essential regulatory mechanism in angiogenesis. We found that uPAR expression on the surface of confluent endothelial cells was down-regulated compared with subconfluent proliferating endothelial cells. Regulation of uPAR expression was most probably affected by extracellular signal-regulated kinase 1/2 (ERK1/2) activation, a downstream signaling event of the VEGF/VEGF-receptor system. Consistently, the receptor-like protein tyrosine phosphatase DEP-1 (density enhanced phosphatase-1/CD148), which is abundantly expressed in confluent endothelial cells, inhibited the VEGF-dependent activation of ERK1/2, leading to down-regulation of uPAR expression. Overexpression of active ERK1 rescued the DEP-1 effect on uPAR. That DEP-1 plays a biologic role in angiogenic endothelial cell behavior was demonstrated in endothelial cell migration, proliferation, and capillary-like tube formation assays in vitro.
Collapse
|
54
|
Petermann A, Haase D, Wetzel A, Balavenkatraman KK, Tenev T, Gührs KH, Friedrich S, Nakamura M, Mawrin C, Böhmer FD. Loss of the protein-tyrosine phosphatase DEP-1/PTPRJ drives meningioma cell motility. Brain Pathol 2010; 21:405-18. [PMID: 21091576 DOI: 10.1111/j.1750-3639.2010.00464.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
DEP-1/PTPRJ is a transmembrane protein-tyrosine phosphatase which has been proposed as a suppressor of epithelial tumors. We have found loss of heterozygosity (LOH) of the PTPRJ gene and loss of DEP-1 protein expression in a subset of human meningiomas. RNAi-mediated suppression of DEP-1 in DEP-1 positive meningioma cell lines caused enhanced motility and colony formation in semi-solid media. Cells devoid of DEP-1 exhibited enhanced signaling of endogenous platelet-derived growth factor (PDGF) receptors, and reduced paxillin phosphorylation upon seeding. Moreover, DEP-1 loss caused diminished adhesion to different matrices, and impaired cell spreading. DEP-1-deficient meningioma cells exhibited invasive growth in an orthotopic xenotransplantation model in nude mice, indicating that elevated motility translates into a biological phenotype in vivo. We propose that negative regulation of PDGF receptor signaling and positive regulation of adhesion signaling by DEP-1 cooperate in inhibition of meningioma cell motility, and possibly tumor invasiveness. These phenotypes of DEP-1 loss reveal functions of DEP-1 in adherent cells, and may be more generally relevant for tumorigenesis.
Collapse
Affiliation(s)
- Astrid Petermann
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Jena, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Motono C, Nakata J, Koike R, Shimizu K, Shirota M, Amemiya T, Tomii K, Nagano N, Sakaya N, Misoo K, Sato M, Kidera A, Hiroaki H, Shirai T, Kinoshita K, Noguchi T, Ota M. SAHG, a comprehensive database of predicted structures of all human proteins. Nucleic Acids Res 2010; 39:D487-93. [PMID: 21051360 PMCID: PMC3013665 DOI: 10.1093/nar/gkq1057] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Most proteins from higher organisms are known to be multi-domain proteins and contain substantial numbers of intrinsically disordered (ID) regions. To analyse such protein sequences, those from human for instance, we developed a special protein-structure-prediction pipeline and accumulated the products in the Structure Atlas of Human Genome (SAHG) database at http://bird.cbrc.jp/sahg. With the pipeline, human proteins were examined by local alignment methods (BLAST, PSI-BLAST and Smith–Waterman profile–profile alignment), global–local alignment methods (FORTE) and prediction tools for ID regions (POODLE-S) and homology modeling (MODELLER). Conformational changes of protein models upon ligand-binding were predicted by simultaneous modeling using templates of apo and holo forms. When there were no suitable templates for holo forms and the apo models were accurate, we prepared holo models using prediction methods for ligand-binding (eF-seek) and conformational change (the elastic network model and the linear response theory). Models are displayed as animated images. As of July 2010, SAHG contains 42 581 protein-domain models in approximately 24 900 unique human protein sequences from the RefSeq database. Annotation of models with functional information and links to other databases such as EzCatDB, InterPro or HPRD are also provided to facilitate understanding the protein structure-function relationships.
Collapse
Affiliation(s)
- Chie Motono
- Computational Biology Research Center, National Institute of Advanced Industrial Science and Technology, Tokyo 135-0064, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Seo HY, Cho SY. PTPε Represses LPS-Mediated TNF-α Induction in RAW264.7 Cells by Inducing Dephosphorylation of p38. B KOREAN CHEM SOC 2010. [DOI: 10.5012/bkcs.2010.31.8.2419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
57
|
Matozaki T, Murata Y, Mori M, Kotani T, Okazawa H, Ohnishi H. Expression, localization, and biological function of the R3 subtype of receptor-type protein tyrosine phosphatases in mammals. Cell Signal 2010; 22:1811-7. [PMID: 20633639 DOI: 10.1016/j.cellsig.2010.07.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2010] [Accepted: 07/06/2010] [Indexed: 11/26/2022]
Abstract
The R3 subtype of receptor-type protein tyrosine phosphatases (RPTPs) includes VE-PTP, DEP-1, PTPRO, and SAP-1. All of these enzymes share a similar structure, with a single catalytic domain and putative tyrosine phosphorylation sites in the cytoplasmic region and fibronectin type III-like domains in the extracellular region. The expression of each R3 RPTP is largely restricted to a single or limited number of cell types, with VE-PTP and DEP-1 being expressed in endothelial or hematopoietic cells, PTPRO in neurons and in podocytes of the renal glomerulus, and SAP-1 in gastrointestinal epithelial cells. In addition, these RPTPs are localized specifically at the apical surface of polarized cells. The structure, expression, and localization of the R3 RPTPs suggest that they perform tissue-specific functions and that they might act through a common mechanism that includes activation of Src family kinases. In this review, we describe recent insights into R3-subtype RPTPs, particularly those of mammals.
Collapse
Affiliation(s)
- Takashi Matozaki
- Laboratory of Biosignal Sciences, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-Machi, Maebashi, Gunma 371-8512, Japan.
| | | | | | | | | | | |
Collapse
|
58
|
Murata Y, Mori M, Kotani T, Supriatna Y, Okazawa H, Kusakari S, Saito Y, Ohnishi H, Matozaki T. Tyrosine phosphorylation of R3 subtype receptor-type protein tyrosine phosphatases and their complex formations with Grb2 or Fyn. Genes Cells 2010; 15:513-24. [PMID: 20398064 DOI: 10.1111/j.1365-2443.2010.01398.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Post-translational modification of protein tyrosine phosphatases (PTPs) is implicated in functional modulation of these enzymes. Stomach cancer-associated protein tyrosine phosphatase-1 (SAP-1), as well as protein tyrosine phosphatase receptor type O (PTPRO) and vascular endothelial-protein tyrosine phosphatase (VE-PTP) are receptor-type PTPs (RPTPs), which belong to the R3 subtype RPTP family. Here, we have shown that the carboxyl (COOH)-terminal region of SAP-1 undergoes tyrosine phosphorylation by the treatment with a PTP inhibitor. Src family kinases are important for the tyrosine phosphorylation of SAP-1. Either Grb2 or Fyn, through their Src homology-2 domains, bound to the tyrosine-phosphorylated SAP-1. Moreover, both PTPRO and VE-PTP underwent tyrosine phosphorylation in their COOH-terminal regions. Tyrosine phosphorylation of VE-PTP or PTPRO also promoted their complex formations with Grb2 or Fyn. Forced expression of SAP-1, PTPRO or VE-PTP promoted cell spreading and lamellipodium formation of fibroblasts that expressed an activated form of Ras. In contrast, such effects of non-tyrosine-phosphorylated forms of these RPTPs were markedly smaller than those of wild-type RPTPs. Our results thus suggest that tyrosine phosphorylation of R3 subtype RPTPs promotes their complex formations with Grb2 or Fyn and thus participates in the regulation of cell morphology.
Collapse
Affiliation(s)
- Yoji Murata
- Laboratory of Biosignal Sciences, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-Machi, Maebashi, Gunma 371-8512, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Tarcic G, Boguslavsky SK, Wakim J, Kiuchi T, Liu A, Reinitz F, Nathanson D, Takahashi T, Mischel PS, Ng T, Yarden Y. An unbiased screen identifies DEP-1 tumor suppressor as a phosphatase controlling EGFR endocytosis. Curr Biol 2010; 19:1788-98. [PMID: 19836242 DOI: 10.1016/j.cub.2009.09.048] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 09/15/2009] [Accepted: 09/15/2009] [Indexed: 01/26/2023]
Abstract
BACKGROUND The epidermal growth factor (EGF) stimulates rapid tyrosine phosphorylation of the EGF receptor (EGFR). This event precedes signaling from both the plasma membrane and from endosomes, and it is essential for recruitment of a ubiquitin ligase, CBL, that sorts activated receptors to endosomes and degradation. Because hyperphosphorylation of EGFR is involved in oncogenic pathways, we performed an unbiased screen of small interfering RNA (siRNA) oligonucleotides targeting all human tyrosine phosphatases. RESULTS We report the identification of PTPRK and PTPRJ (density-enhanced phosphatase-1 [DEP-1]) as EGFR-targeting phosphatases. DEP-1 is a tumor suppressor that dephosphorylates and thereby stabilizes EGFR by hampering its ability to associate with the CBL-GRB2 ubiquitin ligase complex. DEP-1 silencing enhanced tyrosine phosphorylation of endosomal EGFRs and, accordingly, increased cell proliferation. In line with functional interactions, EGFR and DEP-1 form physical associations, and EGFR phosphorylates a substrate-trapping mutant of DEP-1. Interestingly, the interactions of DEP-1 and EGFR are followed by physical segregation: whereas EGFR undergoes endocytosis, DEP-1 remains confined to the cell surface. CONCLUSIONS EGFR and DEP-1 physically interact at the cell surface and maintain bidirectional enzyme-substrate interactions, which are relevant to their respective oncogenic and tumor-suppressive functions. These observations highlight the emerging roles of vesicular trafficking in malignant processes.
Collapse
Affiliation(s)
- Gabi Tarcic
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Mita Y, Yasuda Y, Sakai A, Yamamoto H, Toyooka S, Gunduz M, Tanabe S, Naomoto Y, Ouchida M, Shimizu K. Missense polymorphisms of PTPRJ and PTPN13 genes affect susceptibility to a variety of human cancers. J Cancer Res Clin Oncol 2009; 136:249-59. [DOI: 10.1007/s00432-009-0656-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Accepted: 07/28/2009] [Indexed: 12/28/2022]
|
61
|
de Souza Malaspina TS, Zambuzzi WF, dos Santos CX, Campanelli AP, Laurindo FRM, Sogayar MC, Granjeiro JM. A possible mechanism of low molecular weight protein tyrosine phosphatase (LMW-PTP) activity modulation by glutathione action during human osteoblast differentiation. Arch Oral Biol 2009; 54:642-50. [DOI: 10.1016/j.archoralbio.2009.03.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 03/06/2009] [Accepted: 03/22/2009] [Indexed: 10/20/2022]
|
62
|
Sacco F, Tinti M, Palma A, Ferrari E, Nardozza AP, van Huijsduijnen RH, Takahashi T, Castagnoli L, Cesareni G. Tumor suppressor density-enhanced phosphatase-1 (DEP-1) inhibits the RAS pathway by direct dephosphorylation of ERK1/2 kinases. J Biol Chem 2009; 284:22048-22058. [PMID: 19494114 DOI: 10.1074/jbc.m109.002758] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Density-enhanced phosphatase-1 (DEP-1) is a trans-membrane receptor protein-tyrosine phosphatase that plays a recognized prominent role as a tumor suppressor. However, the mechanistic details underlying its function are poorly understood because its primary physiological substrate(s) have not been firmly established. To shed light on the mechanisms underlying the anti-proliferative role of this phosphatase, we set out to identify new DEP-1 substrates by a novel approach based on screening of high density peptide arrays. The results of the array experiment were combined with a bioinformatics filter to identify eight potential DEP-1 targets among the proteins annotated in the MAPK pathway. In this study we show that one of these potential targets, the ERK1/2, is indeed a direct DEP-1 substrate in vivo. Pulldown and in vitro dephosphorylation assays confirmed our prediction and demonstrated an overall specificity of DEP-1 in targeting the phosphorylated tyrosine 204 of ERK1/2. After epidermal growth factor stimulation, the phosphorylation of the activation loop of ERK1/2 can be modulated by changing the concentration of DEP-1, without affecting the activity of the upstream kinase MEK. In addition, we show that DEP-1 contains a KIM-like motif to recruit ERK1/2 proteins by a docking mechanism mediated by the common docking domain in ERK1/2. ERK proteins that are mutated in the conserved docking domain become insensitive to DEP-1 de-phosphorylation. Overall this study provides novel insights into the anti-proliferative role of this phosphatase and proposes a new mechanism that may also be relevant for the regulation of density-dependent growth inhibition.
Collapse
Affiliation(s)
- Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Michele Tinti
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Anita Palma
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Emanuela Ferrari
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Aurelio P Nardozza
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | | | - Takamune Takahashi
- Nephrology Division and Center for Vascular Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy; Istituto di Ricovero e Cura a Carattere Scientifico Fondazione Santa Lucia, 00143 Rome, Italy
| |
Collapse
|
63
|
Miguet L, Béchade G, Fornecker L, Zink E, Felden C, Gervais C, Herbrecht R, van Dorsselaer A, Mauvieux L, Sanglier-Cianferani S. Proteomic Analysis of Malignant B-Cell Derived Microparticles Reveals CD148 as a Potentially Useful Antigenic Biomarker for Mantle Cell Lymphoma Diagnosis. J Proteome Res 2009; 8:3346-54. [DOI: 10.1021/pr801102c] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Laurent Miguet
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC-DSA, UDS, CNRS, UMR7178, ECPM 25 rue Becquerel, 67087 Strasbourg, France, Laboratoire d’Hématologie cellulaire EA 3948, Institut d’Hématologie, Strasbourg, France, and Pôle d’Onco-Hématologie - Hôpitaux Universitaires de Strasbourg, France
| | - Guillaume Béchade
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC-DSA, UDS, CNRS, UMR7178, ECPM 25 rue Becquerel, 67087 Strasbourg, France, Laboratoire d’Hématologie cellulaire EA 3948, Institut d’Hématologie, Strasbourg, France, and Pôle d’Onco-Hématologie - Hôpitaux Universitaires de Strasbourg, France
| | - Luc Fornecker
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC-DSA, UDS, CNRS, UMR7178, ECPM 25 rue Becquerel, 67087 Strasbourg, France, Laboratoire d’Hématologie cellulaire EA 3948, Institut d’Hématologie, Strasbourg, France, and Pôle d’Onco-Hématologie - Hôpitaux Universitaires de Strasbourg, France
| | - Estelle Zink
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC-DSA, UDS, CNRS, UMR7178, ECPM 25 rue Becquerel, 67087 Strasbourg, France, Laboratoire d’Hématologie cellulaire EA 3948, Institut d’Hématologie, Strasbourg, France, and Pôle d’Onco-Hématologie - Hôpitaux Universitaires de Strasbourg, France
| | - Claire Felden
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC-DSA, UDS, CNRS, UMR7178, ECPM 25 rue Becquerel, 67087 Strasbourg, France, Laboratoire d’Hématologie cellulaire EA 3948, Institut d’Hématologie, Strasbourg, France, and Pôle d’Onco-Hématologie - Hôpitaux Universitaires de Strasbourg, France
| | - Carine Gervais
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC-DSA, UDS, CNRS, UMR7178, ECPM 25 rue Becquerel, 67087 Strasbourg, France, Laboratoire d’Hématologie cellulaire EA 3948, Institut d’Hématologie, Strasbourg, France, and Pôle d’Onco-Hématologie - Hôpitaux Universitaires de Strasbourg, France
| | - Raoul Herbrecht
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC-DSA, UDS, CNRS, UMR7178, ECPM 25 rue Becquerel, 67087 Strasbourg, France, Laboratoire d’Hématologie cellulaire EA 3948, Institut d’Hématologie, Strasbourg, France, and Pôle d’Onco-Hématologie - Hôpitaux Universitaires de Strasbourg, France
| | - Alain van Dorsselaer
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC-DSA, UDS, CNRS, UMR7178, ECPM 25 rue Becquerel, 67087 Strasbourg, France, Laboratoire d’Hématologie cellulaire EA 3948, Institut d’Hématologie, Strasbourg, France, and Pôle d’Onco-Hématologie - Hôpitaux Universitaires de Strasbourg, France
| | - Laurent Mauvieux
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC-DSA, UDS, CNRS, UMR7178, ECPM 25 rue Becquerel, 67087 Strasbourg, France, Laboratoire d’Hématologie cellulaire EA 3948, Institut d’Hématologie, Strasbourg, France, and Pôle d’Onco-Hématologie - Hôpitaux Universitaires de Strasbourg, France
| | - Sarah Sanglier-Cianferani
- Laboratoire de Spectrométrie de Masse Bio-Organique, IPHC-DSA, UDS, CNRS, UMR7178, ECPM 25 rue Becquerel, 67087 Strasbourg, France, Laboratoire d’Hématologie cellulaire EA 3948, Institut d’Hématologie, Strasbourg, France, and Pôle d’Onco-Hématologie - Hôpitaux Universitaires de Strasbourg, France
| |
Collapse
|
64
|
Sallee JL, Burridge K. Density-enhanced phosphatase 1 regulates phosphorylation of tight junction proteins and enhances barrier function of epithelial cells. J Biol Chem 2009; 284:14997-5006. [PMID: 19332538 DOI: 10.1074/jbc.m901901200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cell-cell adhesion is a dynamic process that can activate multiple signaling pathways. These signaling pathways can be regulated through reversible tyrosine phosphorylation events. The level of tyrosine phosphorylation of junctional proteins reflects the balance between protein-tyrosine kinase and protein-tyrosine phosphatase activity. The receptor-tyrosine phosphatase DEP-1 (CD148/PTP-eta) has been implicated in cell growth and differentiation as well as in regulating phosphorylation of junctional proteins. However, the role of DEP-1 in regulating tight junction phosphorylation and the integrity of cell-cell junctions is still under investigation. In this study, we used a catalytically dead substrate-trapping mutant of DEP-1 to identify potential substrates at cell-cell junctions. We have shown that in epithelial cells the trapping mutant of DEP-1 interacts with the tight junction proteins occludin and ZO-1 in a tyrosine phosphorylation-dependent manner. In contrast, PTP-PEST, Shp2, and PTPmu did not interact with these proteins, suggesting that the interaction of DEP-1 with occludin and ZO-1 is specific. In addition, occludin and ZO-1 were dephosphorylated by DEP-1 but not these other phosphatases in vitro. Overexpression of DEP-1 increased barrier function as measured by transepithelial electrical resistance and also reduced paracellular flux of fluorescein isothiocyanate-dextran following a calcium switch. Reduced DEP-1 expression by small interfering RNA had a small but significant increase in junction permeability. These data suggest that DEP-1 can modify the phosphorylation state of tight junction proteins and play a role in regulating permeability.
Collapse
Affiliation(s)
- Jennifer L Sallee
- Department of Cell and Developmental Biology and Lineberger Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | |
Collapse
|
65
|
Dave RK, Hume DA, Elsegood C, Kellie S. CD148/DEP-1 association with areas of cytoskeletal organisation in macrophages. Exp Cell Res 2009; 315:1734-44. [PMID: 19268662 DOI: 10.1016/j.yexcr.2009.02.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 02/15/2009] [Accepted: 02/17/2009] [Indexed: 01/04/2023]
Abstract
In macrophages, tyrosine phosphorylation regulates many signalling pathways leading to growth, differentiation, activation, phagocytosis and adhesion. Protein tyrosine phosphatases (PTPs) represent a biochemical counterbalance to the activity of protein tyrosine kinases, thus regulating the dynamic phosphorylation state of a cell. CD148 is a receptor PTP that is highly expressed in macrophages and is further regulated by pro-inflammatory stimuli. CD148 is normally localised to the plasma membrane of macrophages. Following stimulation with CSF-1 or LPS, there was a re-distribution and concentration of CD148 in areas of membrane ruffling. Treatment of macrophages with anti-CD148 monoclonal antibody inhibited CSF-1-induced macrophage spreading, cytoskeletal re-arrangements and chemotaxis, without affecting cell survival. There were no detectable effects on the CSF-1 receptor-akt signalling pathway. These results are consistent with the hypothesis that CD148 is a regulator of macrophage activity.
Collapse
Affiliation(s)
- Richa K Dave
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Qld, Australia
| | | | | | | |
Collapse
|
66
|
Iuliano R, Raso C, Quintiero A, Pera IL, Pichiorri F, Palumbo T, Palmieri D, Pattarozzi A, Florio T, Viglietto G, Trapasso F, Croce CM, Fusco A. The eighth fibronectin type III domain of protein tyrosine phosphatase receptor J influences the formation of protein complexes and cell localization. J Biochem 2009; 145:377-85. [PMID: 19122201 DOI: 10.1093/jb/mvn175] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Regulation of receptor-type phosphatases can involve the formation of higher-order structures, but the exact role played in this process by protein domains is not well understood. In this study we show the formation of different higher-order structures of the receptor-type phosphatase PTPRJ, detected in HEK293A cells transfected with different PTPRJ expression constructs. In the plasma membrane PTPRJ forms dimers detectable by treatment with the cross-linking reagent BS(3) (bis[sulfosuccinimidyl]suberate). However, other PTPRJ complexes, dependent on the formation of disulfide bonds, are detected by treatment with the oxidant agent H(2)O(2) or by a mutation Asp872Cys, located in the eighth fibronectin type III domain of PTPRJ. A deletion in the eighth fibronectin domain of PTPRJ impairs its dimerization in the plasma membrane and increases the formation of PTPRJ complexes dependent on disulfide bonds that remain trapped in the cytoplasm. The deletion mutant maintains the catalytic activity but is unable to carry out inhibition of proliferation on HeLa cells, achieved by the wild type form, since it does not reach the plasma membrane. Therefore, the intact structure of the eighth fibronectin domain of PTPRJ is critical for its localization in plasma membrane and biological function.
Collapse
Affiliation(s)
- Rodolfo Iuliano
- Dipartimento di Medicina Sperimentale e Clinica, Facoltà di Medicina e Chirurgia, Università di Catanzaro, 88100 Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
New role for the protein tyrosine phosphatase DEP-1 in Akt activation and endothelial cell survival. Mol Cell Biol 2008; 29:241-53. [PMID: 18936167 DOI: 10.1128/mcb.01374-08] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Functional inactivation of the protein tyrosine phosphatase DEP-1 leads to increased endothelial cell proliferation and failure of vessels to remodel and branch. DEP-1 has also been proposed to contribute to the contact inhibition of endothelial cell growth via dephosphorylation of vascular endothelial growth factor receptor 2 (VEGFR2), a mediator of vascular development. However, how DEP-1 regulates VEGF-dependent signaling and biological responses remains ill-defined. We show here that DEP-1 targets tyrosine residues in the VEGFR2 kinase activation loop. Consequently, depletion of DEP-1 results in the increased phosphorylation of all major VEGFR2 autophosphorylation sites, but surprisingly, not in the overall stimulation of VEGF-dependent signaling. The increased phosphorylation of Src on Y529 under these conditions results in impaired Src and Akt activation. This inhibition is similarly observed upon expression of catalytically inactive DEP-1, and coexpression of an active Src-Y529F mutant rescues Akt activation. Reduced Src activity correlates with decreased phosphorylation of Gab1, an adapter protein involved in VEGF-dependent Akt activation. Hypophosphorylated Gab1 is unable to fully associate with phosphatidylinositol 3-kinase, VEGFR2, and VE-cadherin complexes, leading to suboptimal Akt activation and increased cell death. Overall, our results reveal that despite its negative role on global VEGFR2 phosphorylation, DEP-1 is a positive regulator of VEGF-mediated Src and Akt activation and endothelial cell survival.
Collapse
|
68
|
Rodriguez F, Vacaru A, Overvoorde J, den Hertog J. The receptor protein-tyrosine phosphatase, Dep1, acts in arterial/venous cell fate decisions in zebrafish development. Dev Biol 2008; 324:122-30. [PMID: 18835554 DOI: 10.1016/j.ydbio.2008.09.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 09/08/2008] [Accepted: 09/09/2008] [Indexed: 02/07/2023]
Abstract
Dep1 is a transmembrane protein-tyrosine phosphatase (PTP) that is expressed in vascular endothelial cells and has tumor suppressor activity. Mouse models with gene targeted Dep1 either show vascular defects, or do not show any defects at all. We used the zebrafish to investigate the role of Dep1 in early development. The zebrafish genome encodes two highly homologous Dep1 genes, Dep1a and Dep1b. Morpholinos specific for Dep1a and Dep1b induced defects in vasculature, resulting in defective blood circulation. However, Green Fluorescent Protein expression in fli1a::gfp1 transgenic embryos and cdh5 expression, markers of vascular endothelial cells, were normal upon Dep1a- and Dep1b-MO injection. Molecular markers indicated that arterial specification was reduced and venous markers were expanded in Dep1 morphants. Moreover, the Dep1a/Dep1b knockdowns were rescued by inhibition of Phosphatidylinositol-3 kinase (PI3K) and by expression of active Notch and Grl/Hey2. Our results suggest a model in which Dep1 acts upstream in a signaling pathway inhibiting PI3K, resulting in expression of Notch and Grl, thus regulating arterial specification in development.
Collapse
Affiliation(s)
- Fiona Rodriguez
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | | | | | | |
Collapse
|
69
|
Karagyozov L, Godfrey R, Böhmer SA, Petermann A, Hölters S, Ostman A, Böhmer FD. The structure of the 5'-end of the protein-tyrosine phosphatase PTPRJ mRNA reveals a novel mechanism for translation attenuation. Nucleic Acids Res 2008; 36:4443-53. [PMID: 18603590 PMCID: PMC2490741 DOI: 10.1093/nar/gkn391] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Analysis of the human protein-tyrosine phosphatase (PTP) PTPRJ mRNA detected three in-frame AUGs at the 5′-end (starting at nt +14, +191 and +356) with no intervening stop codons. This tandem AUG arrangement is conserved between humans and the mouse and is unique among the genes of the classical PTPs. Until now it was assumed that the principal open reading frame (ORF) starts at AUG356. Our experiments showed that: (i) translation of the mRNA synthesized under the PTPRJ promoter starts predominantly at AUG191, leading to the generation of a 55 amino acid sequence preceding the signal peptide; (ii) the longer form is being likewise correctly processed into mature PTPRJ; (iii) the translation of the region between AUG191 and AUG356 inhibits the overall expression, a feature which depends on the sequence of the encoded peptide. Specifically, a sequence of 13 amino acids containing multiple arginine residues (RRTGWRRRRRRRR) confers the inhibition. In the absence of uORF these previously unrecognized characteristics of the 5′-end of the mRNA present a novel mechanism to suppress, and potentially to regulate translation.
Collapse
Affiliation(s)
- Luchezar Karagyozov
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, Friedrich-Schiller-University Jena, Jena, Germany
| | | | | | | | | | | | | |
Collapse
|
70
|
The tyrosine phosphatase CD148 interacts with the p85 regulatory subunit of phosphoinositide 3-kinase. Biochem J 2008; 413:193-200. [PMID: 18348712 DOI: 10.1042/bj20071317] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
CD148 is a transmembrane tyrosine phosphatase that has been implicated in the regulation of cell growth and transformation. However, the signalling mechanisms of CD148 are incompletely understood. To identify the specific intracellular molecules involved in CD148 signalling, we carried out a modified yeast two-hybrid screening assay. Using the substrate-trapping mutant form of CD148 (CD148 D/A) as bait, we recovered the p85 regulatory subunit of PI3K (phosphoinositide 3-kinase). CD148 D/A, but not catalytically active CD148, interacted with p85 in a phosphorylation-dependent manner in vitro and in intact cells. Growth factor receptor and PI3K activity were also trapped by CD148 D/A via p85 from pervanadate-treated cell lysates. CD148 prominently and specifically dephosphorylated p85 in vitro. Co-expression of CD148 reduced p85 phosphorylation induced by active Src, and attenuated the increases in PI3K activity, yet CD148 did not alter the basal PI3K activity. Finally, CD148 knock-down by siRNA (short interfering RNA) increased PI3K activity on serum stimulation. Taken together, these results demonstrate that CD148 may interact with and dephosphorylate p85 when it is phosphorylated and modulate the magnitude of PI3K activity.
Collapse
|
71
|
Abstract
Protein-tyrosine phosphatases are tightly controlled by various mechanisms, ranging from differential expression in specific cell types to restricted subcellular localization, limited proteolysis, post-translational modifications affecting intrinsic catalytic activity, ligand binding and dimerization. Here, we review the regulatory mechanisms found to control the classical protein-tyrosine phosphatases.
Collapse
|
72
|
Chen WL, Lin CT, Lo HF, Lee JW, Tu IH, Hu FR. The role of protein tyrosine phosphorylation in the cell-cell interactions, junctional permeability and cell cycle control in post-confluent bovine corneal endothelial cells. Exp Eye Res 2007; 85:259-69. [PMID: 17624326 DOI: 10.1016/j.exer.2007.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2006] [Revised: 04/14/2007] [Accepted: 04/25/2007] [Indexed: 12/31/2022]
Abstract
Cell-cell interaction, junctional permeability and contact inhibition are important mechanisms that allow corneal endothelial cells to maintain stable corneal hydration status and also keep these cells in non-proliferative status. Protein tyrosine phosphatases (PTPs) are well known to play an important role in regulating cell-cell contacts, growth and differentiation. Inhibition of PTPs activity by a general PTP inhibitor has been proved to trigger post-confluent rat corneal endothelial cells to reenter cell cycles. In this study, we aimed to evaluate whether protein tyrosine phosphorylation is involved in cell-cell interactions, junctional permeability and cell cycle control in post-confluent, contact inhibited bovine corneal endothelial cells. Confluent cultures of bovine corneal endothelial cells were treated with different concentrations of general phosphatase inhibitor, sodium orthovanadate (vanadate) for several different durations. Protein tyrosine phosphorylation was confirmed by Western blot analysis. Immunocytochemistry was used to evaluate the effect of vanadate on adherens-type junctional proteins by staining of p120, N-cadherin and alpha-catenin. Paracelluar permeability was evaluated by transepithelial electric resistance. The effect of vanadate on cell cycle progression was confirmed by immunostaining of Ki67. Western blot analysis was used to evaluate the expression level of cell-cycle-associated proteins, including PCNA, cyclin D1, cyclin E and cyclin A. Time-dependent effects of vanadate on protein tyrosine phosphorylation were confirmed by Western blot analysis. ICC demonstrated the effect of vanadate on the disruption of p120, N-cadherin and alpha-catenin. Time- and dose-effects of vanadate on the severity of p120 disruption were also found. TER demonstrated the time- and dose-effect of vanadte on paracellular permeability. Although cell-cell junctions can be broken through by vanadate, no significant increase of Ki67 positive cells was found among the control group and all groups with different concentrations/durations of vanadate treatment. Western blot also showed no change of PCNA, cyclin D1, cyclin E and cyclin A after treatment with vanadate. In conclusion, protein tyrosine phosphatase inhibition can induce time-dependent release of cell-cell contacts and increase transepithelial permeability in post-confluent cultures of bovine corneal endothelial cells. However, such phenomenon is not enough to promoted cell cycle progression as seen in rat corneal endothelial cells.
Collapse
Affiliation(s)
- Wei-Li Chen
- Department of Ophthalmology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
73
|
Affiliation(s)
- Tasneem Motiwala
- Department of Molecular and Cellular Biochemistry, The Ohio State University, College of Medicine, Columbus, Ohio 43210, USA
| | | |
Collapse
|
74
|
Matozo HC, Nascimento AS, Santos MAM, Iuliano R, Fusco A, Polikarpov I. Crystallization and preliminary X-ray diffraction analysis of rat protein tyrosine phosphatase eta. Acta Crystallogr Sect F Struct Biol Cryst Commun 2006; 62:923-5. [PMID: 16946481 PMCID: PMC2242866 DOI: 10.1107/s1744309106031058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Accepted: 08/07/2006] [Indexed: 11/10/2022]
Abstract
The rat protein tyrosine phosphatase eta (rPTPeta) is a cysteine-dependent phosphatase which hydrolyzes phosphoester bonds in proteins and other molecules. rPTPeta and its human homologue DEP-1 are involved in neoplastic transformations. Thus, expression of the protein is reduced in all oncogene-transformed thyroid cell lines and is absent in highly malignant thyroid cells. Moreover, consistent with the suggested tumour suppression role of PTPeta, inhibition of the tumorigenic process occurs after its exogenous reconstitution, suggesting that PTPeta might be important for gene therapy of cancers. In this study, the catalytic domain of rPTPeta was produced in Escherichia coli in soluble form and purified to homogeneity. Crystals were obtained by the hanging-drop vapour-diffusion method. Diffraction data were collected to 1.87 A resolution. The crystal belongs to space group P2(1)2(1)2(1), with unit-cell parameters a = 46.46, b = 63.07, c = 111.64 A, and contains one molecule per asymmetric unit.
Collapse
Affiliation(s)
- Huita C Matozo
- Instituto de Física de São Carlos, Departamento de Física e Informática, Universidade de São Paulo, Avenida Trabalhador São Carlense 400, CEP 13566-590 São Carlos, SP, Brazil
| | | | | | | | | | | |
Collapse
|
75
|
Trapasso F, Drusco A, Costinean S, Alder H, Aqeilan RI, Iuliano R, Gaudio E, Raso C, Zanesi N, Croce CM, Fusco A. Genetic ablation of Ptprj, a mouse cancer susceptibility gene, results in normal growth and development and does not predispose to spontaneous tumorigenesis. DNA Cell Biol 2006; 25:376-82. [PMID: 16792508 DOI: 10.1089/dna.2006.25.376] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ptprj is a ubiquitously expressed murine gene encoding a receptor-type protein tyrosine phosphatase, which has recently been proposed as a candidate gene on the locus Scc1 for colon cancer susceptibility. It has been demonstrated that PTPRJ, the human homologue of Ptprj, is involved in the control of cell growth and adhesion, being furthermore altered in several types of cancer including mammary, thyroid, lung, colon, and pancreatic cancers. To investigate the biological functions of Ptprj, we have generated mice deficient in this receptor protein tyrosine phosphatase. Ptprj-deficient mice are viable, fertile, and show no gross anatomical alterations. Furthermore, neither changes in life span nor spontaneous tumor appearance were observed in Ptprj-null mice. Our results indicate that Ptprj is dispensable for normal growth and development in mice.
Collapse
Affiliation(s)
- Francesco Trapasso
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia di Catanzaro, Campus Germaneto, Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Alemà S, Salvatore AM. p120 catenin and phosphorylation: Mechanisms and traits of an unresolved issue. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1773:47-58. [PMID: 16904204 DOI: 10.1016/j.bbamcr.2006.06.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Revised: 06/02/2006] [Accepted: 06/06/2006] [Indexed: 01/11/2023]
Abstract
p120 catenin is a scaffold protein that interacts with cadherin cytoplasmic domain and acts as a crucial component of the signalling that regulates the cycle of adherens junction formation and disassembly. Here, we review the nature of stimuli that modulate p120ctn function and are translated as serine/threonine and tyrosine phosphorylation events at this multisite substrate for a variety of protein kinases. We also highlight recent findings that tentatively link phosphorylation of p120ctn to its role as a signal integrator capable to influence the state of the cadherin adhesive bond, the cytoskeleton and cell motility.
Collapse
Affiliation(s)
- Stefano Alemà
- Istituto di Biologia Cellulare, CNR, 00016 Monterotondo, Italy
| | | |
Collapse
|
77
|
Anders L, Mertins P, Lammich S, Murgia M, Hartmann D, Saftig P, Haass C, Ullrich A. Furin-, ADAM 10-, and gamma-secretase-mediated cleavage of a receptor tyrosine phosphatase and regulation of beta-catenin's transcriptional activity. Mol Cell Biol 2006; 26:3917-34. [PMID: 16648485 PMCID: PMC1489012 DOI: 10.1128/mcb.26.10.3917-3934.2006] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Several receptor protein tyrosine phosphatases (RPTPs) are cell adhesion molecules involved in homophilic interactions, suggesting that RPTP outside-in signaling is coupled to cell contact formation. However, little is known about the mechanisms by which cell density regulates RPTP function. We show that the MAM family prototype RPTPkappa is cleaved by three proteases: furin, ADAM 10, and gamma-secretase. Cell density promotes ADAM 10-mediated cleavage and shedding of RPTPkappa. This is followed by gamma-secretase-dependent intramembrane proteolysis of the remaining transmembrane part to release the phosphatase intracellular portion (PIC) from the membrane, thereby allowing its translocation to the nucleus. When cells were treated with leptomycin B, a nuclear export inhibitor, PIC accumulated in nuclear bodies. PIC is an active protein tyrosine phosphatase that binds to and dephosphorylates beta-catenin, an RPTPkappa substrate. The expression of RPTPkappa suppresses beta-catenin's transcriptional activity, whereas the expression of PIC increases it. Notably, this increase required the phosphatase activity of PIC. Thus, both isoforms have acquired opposing roles in the regulation of beta-catenin signaling. We also found that RPTPmu, another MAM family member, undergoes gamma-secretase-dependent processing. Our results identify intramembrane proteolysis as a regulatory switch in RPTPkappa signaling and implicate PIC in the activation of beta-catenin-mediated transcription.
Collapse
Affiliation(s)
- Lars Anders
- Department of Molecular Biology, Max Planck Institute of Biochemistry, Martinsried, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Balavenkatraman KK, Jandt E, Friedrich K, Kautenburger T, Pool-Zobel BL, Ostman A, Böhmer FD. DEP-1 protein tyrosine phosphatase inhibits proliferation and migration of colon carcinoma cells and is upregulated by protective nutrients. Oncogene 2006; 25:6319-24. [PMID: 16682945 DOI: 10.1038/sj.onc.1209647] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The transmembrane protein-tyrosine phosphatase (PTP) DEP-1 (density-enhanced phosphatase) is a candidate tumor suppressor in the colon epithelium. We have explored the function of DEP-1 in colon epithelial cells by inducible re-expression in a DEP-1-deficient human colon cancer cell line. Density-enhanced phosphatase-1 re-expression led to profound inhibition of cell proliferation and cell migration, and was associated with cytoskeletal rearrangements. These effects were dependent on the PTP activity of DEP-1 as they were not observed with cells expressing the catalytically inactive DEP-1 C1239S variant. shRNA-mediated suppression of DEP-1 in a colon epithelial cell line with high endogenous DEP-1 levels enhanced proliferation, further supporting the antiproliferative function of DEP-1. Nutrients, which are considered to be chemoprotective with respect to colon cancer development, including butyrate, green tea and apple polyphenols, had the capacity to elevate transcription of endogenous DEP-1 mRNA and expression of DEP-1 protein. Upregulation of DEP-1 expression, and in turn inhibition of cell growth and migration may present a previously unrecognized mechanism of chemoprevention by nutrients.
Collapse
MESH Headings
- Adenocarcinoma/enzymology
- Adenocarcinoma/pathology
- Adenoma/enzymology
- Adenoma/pathology
- Anticarcinogenic Agents/pharmacology
- Butyrates/pharmacology
- Cell Division/drug effects
- Cell Line, Tumor/cytology
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/enzymology
- Cell Movement/drug effects
- Cells, Cultured/cytology
- Cells, Cultured/drug effects
- Cells, Cultured/enzymology
- Chemokine CXCL12
- Chemokines, CXC/pharmacology
- Colon/cytology
- Colon/enzymology
- Colonic Neoplasms/enzymology
- Colonic Neoplasms/pathology
- Down-Regulation
- Enzyme Induction/drug effects
- Epithelial Cells/cytology
- Epithelial Cells/drug effects
- Epithelial Cells/enzymology
- Flavonoids/pharmacology
- Humans
- Lysophospholipids/pharmacology
- Malus/chemistry
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Phenols/pharmacology
- Plant Extracts/pharmacology
- Polyphenols
- Protein Phosphatase 1
- Protein Tyrosine Phosphatases/biosynthesis
- Protein Tyrosine Phosphatases/genetics
- Protein Tyrosine Phosphatases/physiology
- RNA Interference
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- RNA, Small Interfering/pharmacology
- Receptor-Like Protein Tyrosine Phosphatases, Class 3
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- Tea/chemistry
- Transcription, Genetic/drug effects
- Transfection
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- K K Balavenkatraman
- Institute of Molecular Cell Biology, Medical Faculty, Friedrich Schiller University, Jena, Germany
| | | | | | | | | | | | | |
Collapse
|
79
|
Takahashi T, Takahashi K, Mernaugh RL, Tsuboi N, Liu H, Daniel TO. A monoclonal antibody against CD148, a receptor-like tyrosine phosphatase, inhibits endothelial-cell growth and angiogenesis. Blood 2006; 108:1234-42. [PMID: 16597593 PMCID: PMC1895872 DOI: 10.1182/blood-2005-10-4296] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Angiogenesis contributes to a wide range of neoplastic, ischemic, and inflammatory disorders. Definition of the intrinsic molecular controls in angiogenic vessel growth promises novel therapeutic approaches for angiogenesis-related diseases. CD148 (also named DEP-1/PTP eta) is a receptor-like protein tyrosine phosphatase that is abundantly expressed in vascular endothelial cells. To explore a role of CD148 in endothelial vessel formation, we generated a monoclonal antibody, Ab1, against the ectodomain sequence of CD148 and examined its effects on endothelial-cell growth and vessel formation. Here we report that a bivalent, but not a monovalent, form of the Ab1 antibody inhibits endothelial-cell growth and blocks angiogenesis in mouse cornea in vivo. We further demonstrate that (1) bivalent Ab1 arrests cell-cycle progression of CD148-transfected CHO cells at G(0)/G(1) phase, (2) coexpression of catalytically inactive CD148 mutants attenuates the Ab1-cell growth inhibition, and (3) bivalent Ab1 suppresses phosphorylation of ERK1/2 kinases and Met tyrosine kinase as activated CD148 does, with an increase in CD148-associated tyrosine phosphatase activity. Taken together, these findings demonstrate that Ab1-induced ectodomain oligomerization arrests endothelial-cell growth through catalytic activity of the CD148 cytoplasmic domain. The present study defines CD148 as a valuable molecular target for antiangiogenesis therapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- CHO Cells
- Cornea/blood supply
- Cornea/immunology
- Cornea/metabolism
- Cornea/pathology
- Cricetinae
- Cricetulus
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Enzyme Inhibitors/immunology
- Enzyme Inhibitors/pharmacology
- G1 Phase/drug effects
- G1 Phase/genetics
- G1 Phase/immunology
- Humans
- Inflammation/drug therapy
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/metabolism
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/immunology
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/immunology
- Mitogen-Activated Protein Kinase 3/metabolism
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/metabolism
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/metabolism
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- Protein Processing, Post-Translational/genetics
- Protein Processing, Post-Translational/immunology
- Protein Structure, Tertiary/genetics
- Protein Tyrosine Phosphatases/antagonists & inhibitors
- Protein Tyrosine Phosphatases/genetics
- Protein Tyrosine Phosphatases/immunology
- Proto-Oncogene Proteins c-met/genetics
- Proto-Oncogene Proteins c-met/immunology
- Proto-Oncogene Proteins c-met/metabolism
- Receptor-Like Protein Tyrosine Phosphatases, Class 3
- Resting Phase, Cell Cycle/drug effects
- Resting Phase, Cell Cycle/genetics
- Resting Phase, Cell Cycle/immunology
Collapse
Affiliation(s)
- Takamune Takahashi
- Vanderbilt University Medical Center, Division of Nephrology, Nashville, TN 37232, USA.
| | | | | | | | | | | |
Collapse
|
80
|
Sallee JL, Wittchen ES, Burridge K. Regulation of cell adhesion by protein-tyrosine phosphatases: II. Cell-cell adhesion. J Biol Chem 2006; 281:16189-92. [PMID: 16497667 DOI: 10.1074/jbc.r600003200] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cell-cell adhesion is critical to the development and maintenance of multicellular organisms. The stability of many adhesions is regulated by protein tyrosine phosphorylation of cell adhesion molecules and their associated components, with high levels of phosphorylation promoting disassembly. The level of tyrosine phosphorylation reflects the balance between protein-tyrosine kinase and protein-tyrosine phosphatase activity. Many protein-tyrosine phosphatases associate with the cadherin-catenin complex, directly regulating the phosphorylation of these proteins, thereby affecting their interactions and the integrity of cell-cell junctions. Tyrosine phosphatases can also affect cell-cell adhesions indirectly by regulating the signaling pathways that control the activities of Rho family G proteins. In addition, receptor-type tyrosine phosphatases can mediate outside-in signaling through both ligand binding and dimerization of their extracellular domains. This review will discuss the role of protein-tyrosine phosphatases in cell-cell interactions, with an emphasis on cadherin-mediated adhesions.
Collapse
Affiliation(s)
- Jennifer L Sallee
- Department of Cell and Developmental Biology and Lineberger Cancer Center, University of North Carolina, Chapel Hill, NC 27599-7295, USA
| | | | | |
Collapse
|
81
|
Xu Y, Tan LJ, Grachtchouk V, Voorhees JJ, Fisher GJ. Receptor-type protein-tyrosine phosphatase-kappa regulates epidermal growth factor receptor function. J Biol Chem 2005; 280:42694-700. [PMID: 16263724 DOI: 10.1074/jbc.m507722200] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Epidermal growth factor receptor (EGFR), the prototypic receptor protein tyrosine kinase, is a major regulator of growth and survival for many epithelial cell types. We report here that receptor-type protein-tyrosine phosphatase-kappa (RPTP-kappa) dephosphorylates EGFR and thereby regulates its function in human keratinocytes. Protein-tyrosine phosphatase (PTP) inhibitors induced EGFR tyrosine phosphorylation in intact primary human keratinocytes and cell-free membrane preparations. Five highly expressed RPTPs (RPTP-beta, delta, kappa, mu, and xi) were functionally analyzed in a Chinese hamster ovary (CHO) cell-based expression system. Full-length human EGFR expressed in CHO cells, which lack endogenous EGFR, displayed high basal (i.e. in the absence of ligand) tyrosine phosphorylation. Co-expression of RPTP-kappa, but not other RPTPs, specifically reduced basal EGFR tyrosine phosphorylation. RPTP-kappa also reduced epidermal growth factor-dependent EGFR tyrosine phosphorylation in CHO cells. Purified RPTP-kappa preferentially dephosphorylated EGFR tyrosines 1068 and 1173 in vitro. Overexpression of wild-type or catalytically inactive RPTP-kappa reduced or enhanced, respectively, basal and EGF-induced EGFR tyrosine phosphorylation in human keratinocytes. Furthermore, siRNA-mediated knockdown of RPTP-kappa increased basal and EGF-stimulated EGFR tyrosine phosphorylation and augmented downstream Erk activation in human keratinocytes. RPTP-kappa levels increased in keratinocytes as cells reached confluency, and overexpression of RPTP-kappa in subconfluent keratinocytes reduced keratinocyte proliferation. Taken together, the above data indicate that RPTP-kappa is a key regulator of EGFR tyrosine phosphorylation and function in human keratinocytes.
Collapse
Affiliation(s)
- Yiru Xu
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|
82
|
Berset TA, Hoier EF, Hajnal A. The C. elegans homolog of the mammalian tumor suppressor Dep-1/Scc1 inhibits EGFR signaling to regulate binary cell fate decisions. Genes Dev 2005; 19:1328-40. [PMID: 15901674 PMCID: PMC1142556 DOI: 10.1101/gad.333505] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Protein phosphorylation by kinases and the subsequent dephosphorylation by phosphatases are key mechanisms that regulate intracellular signal transduction during development. Here, we report the identification of the receptor protein tyrosine phosphatase DEP-1 as a negative regulator of the Caenorhabditis elegans EGF receptor. DEP-1 amplifies in the developing vulva and the excretory system the small differences in the amount of EGF signal received by equivalent precursor cells to achieve binary cell fate decisions. During vulval development, DEP-1 inhibits EGFR signaling in the secondary cell lineage in parallel with the NOTCH-mediated lateral inhibition, while EGFR signaling simultaneously down-regulates DEP-1 and NOTCH expression in the primary cell lineage. This regulatory network of inhibitors results in the full activation of the EGFR/RAS/MAPK pathway in the primary vulval cells and at the same time keeps the EGFR/RAS/MAPK pathway inactive in the adjacent secondary cells. Mammalian Dep-1/Scc1 functions as a tumor-suppressor gene in the intestinal epithelium. Thus, mutations in human Dep-1 may promote tumor formation through a hyperactivation of the EGF receptor.
Collapse
|
83
|
Sui XF, Kiser TD, Hyun SW, Angelini DJ, Del Vecchio RL, Young BA, Hasday JD, Romer LH, Passaniti A, Tonks NK, Goldblum SE. Receptor protein tyrosine phosphatase micro regulates the paracellular pathway in human lung microvascular endothelia. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1247-58. [PMID: 15793303 PMCID: PMC1602370 DOI: 10.1016/s0002-9440(10)62343-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The pulmonary vascular endothelial paracellular pathway and zonula adherens (ZA) integrity are regulated, in part, through protein tyrosine phosphorylation. ZA-associated protein tyrosine phosphatase (PTP)s are thought to counterregulate tyrosine phosphorylation events within the ZA multiprotein complex. One such receptor PTP, PTPmu, is highly expressed in lung tissue and is almost exclusively restricted to the endothelium. We therefore studied whether PTPmu, in pulmonary vascular endothelia, associates with and/or regulates both the tyrosine phosphorylation state of vascular endothelial (VE)-cadherin and the paracellular pathway. PTPmu was expressed in postconfluent human pulmonary artery and lung microvascular endothelial cells (ECs) where it was almost exclusively restricted to EC-EC boundaries. In human lung microvascular ECs, knockdown of PTPmu through RNA interference dramatically impaired barrier function. In immortalized human microvascular ECs, overexpression of wild-type PTPmu enhanced barrier function. PTPmu-VE-cadherin interactions were demonstrated through reciprocal co-immunoprecipitation assays and co-localization with double-label fluorescence microscopy. When glutathione S-transferase-PTPmu was incubated with purified recombinant VE-cadherin, and when glutathione S-transferase-VE-cadherin was incubated with purified recombinant PTPmu, PTPmu directly bound to VE-cadherin. Overexpression of wild-type PTPmu decreased tyrosine phosphorylation of VE-cadherin. Therefore, PTPmu is expressed in human pulmonary vascular endothelia where it directly binds to VE-cadherin and regulates both the tyrosine phosphorylation state of VE-cadherin and barrier integrity.
Collapse
Affiliation(s)
- Xiu Fen Sui
- Department of Medicine and Pathology, Division of Infectious Diseases and Pulmonary Medicine, Mucosal Biology Research Center, University of Maryland School of Medicine, 22 Penn St., Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Santos MAM, Santos SM, Matozo HC, Portugal RV, Iuliano R, Fusco A, Polikarpov I. Expression, purification, and characterization of rat protein tyrosine phosphatase η catalytic domain. Protein Expr Purif 2005; 41:113-20. [PMID: 15802228 DOI: 10.1016/j.pep.2005.01.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2004] [Revised: 01/23/2005] [Indexed: 02/07/2023]
Abstract
Receptor-like protein tyrosine phosphatases generally contain one or two conserved intracellular catalytic domains with a conserved sequence motif ([I/V]HCXAGXXR[S/T]G), a single transmembrane domain, and an external highly variable part. Here, we describe cloning of the intracellular catalytic domain of the rat protein tyrosine phosphatase eta (rPTPetaCD) into pET28a(+) vector, its expression in Escherichia coli, purification and initial characterization. The purification of His6-tagged rPTPetaCD to near homogeneity was achieved by a combination of affinity and size exclusion chromatography. The His-tag was subsequently removed by thrombin digestion. PhastGel IEF electrophoresis demonstrated that the isoelectric point of this 41 kDa His6-tag free recombinant protein was 7.3, which is just slightly higher than the theoretically predicted value of 7.2. To assess the functionality of the rPTPetaCD we used the pNPP hydrolysis assay and observed that the enzyme has a specific activity of 9 nmol/min/mug. The secondary structure and stability of the recombinant protein was also analyzed by circular dichroism and fluorescence spectroscopy. In summary, the rPTPetaCD is stable at 18 degrees C, properly folded, and fully active, which makes it a suitable candidate for structural and functional studies.
Collapse
Affiliation(s)
- Maria A M Santos
- Instituto de Física de São Carlos, Departamento de Física e Informática, Universidade de São Paulo, Avenida Trabalhador São Carlense, 400, CEP 13566-590 São Carlos, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
85
|
Pera IL, Iuliano R, Florio T, Susini C, Trapasso F, Santoro M, Chiariotti L, Schettini G, Viglietto G, Fusco A. The rat tyrosine phosphatase η increases cell adhesion by activating c-Src through dephosphorylation of its inhibitory phosphotyrosine residue. Oncogene 2005; 24:3187-95. [PMID: 15735685 DOI: 10.1038/sj.onc.1208510] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The expression of the receptor protein tyrosine phosphatase r-PTPeta is drastically reduced in rat and human malignant thyroid cells, whereas its restoration reverts the neoplastic phenotype of retrovirally transformed rat thyroid cells. Moreover, reduced levels and loss of heterozygosity of DEP-1, the human homolog of r-PTPeta, have been found in many human neoplasias. Here, we report that the r-PTPeta protein binds to c-Src in living cells and dephosphorylates the c-Src inhibitory tyrosine phosphorylation site (Tyr 529), thereby increasing c-Src tyrosine kinase activity in malignant rat thyroid cells stably transfected with r-PTPeta. Tyrosine phosphorylation of focal adhesion kinase (FAK) and paxillin was enhanced in r-PTPeta-expressing cells. This was associated with increased adhesion of malignant r-PTPeta-transfected thyroid cells vs both untransfected cells and cells stably transfected with an inactive r-PTPeta mutant. Treatment of rat thyroid cells with the c-Src inhibitor PP2 decreased cell adhesion to a higher extent in r-PTPeta-transfected cells than in mock-transfected or stably transfected cells with the inactive r-PTPeta mutant, indicating that r-PTPeta regulates cell-substratum adhesion by activating c-Src. Interestingly, the extent of both c-Src dephosphorylation at Tyr 529, FAK and paxillin phosphorylation, and the increased cell adhesion were associated with the degree of r-PTPeta expression.
Collapse
Affiliation(s)
- Ilaria Le Pera
- Department of Experimental and Clinical Medicine, Medical School of Catanzaro, 'Magna Graecia' University of Catanzaro, 88100 Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Schiller KR, Mauro LJ. Tyrosine phosphatases as regulators of skeletal development and metabolism. J Cell Biochem 2005; 96:262-77. [PMID: 16052478 DOI: 10.1002/jcb.20515] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The protein tyrosine kinases (PTK) and the protein tyrosine phosphatases (PTPs) are enzymes which play an integral role in tyrosine phosphorylation-dependent signaling cascades. By catalyzing the phosphorylation and dephosphorylation of cellular proteins, these enzymes direct the steady-state levels of specific phosphoproteins and ultimately dictate the functional state of all cells. The importance of this type of signaling in the skeleton is accepted but poorly understood. The contribution of the PTKs to signaling events in bone has been well studied but, in contrast, the regulation by PTPs is poorly defined. The recent identification of 107 genes within the human genome which encode members of the PTP superfamily emphasizes the need to consider the importance of these proteins in skeletal tissue. In this prospective, we will summarize the present state of our knowledge regarding the function of this enzyme superfamily, illustrating its relevance to the development and maintenance of the skeleton and highlighting future directions that should improve our understanding of these critical signaling molecules.
Collapse
Affiliation(s)
- Katherine R Schiller
- Department of Animal Science, Physiology & Growth Division, University of Minnesota, Minnesota 55108, USA
| | | |
Collapse
|
87
|
Del Vecchio RL, Tonks NK. The Conserved Immunoglobulin Domain Controls the Subcellular Localization of the Homophilic Adhesion Receptor Protein-tyrosine Phosphatase μ. J Biol Chem 2005; 280:1603-12. [PMID: 15491993 DOI: 10.1074/jbc.m410181200] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The receptor protein-tyrosine phosphatase mu (PTPmu) is a homophilic adhesion protein thought to regulate cell-cell adhesion in the vascular endothelium through dephosphorylation of cell junction proteins. In subconfluent cell cultures, PTPmu resides in an intracellular membrane pool; however, as culture density increases and cell contacts form, the phosphatase localizes to sites of cell-cell contact, and its expression level increases. These characteristics of PTPmu, which are consistent with a role in cell-cell adhesion, suggest that control of subcellular localization is an important mechanism to regulate the function of this phosphatase. To gain a better understanding of how PTPmu is regulated, we examined the importance of the conserved immunoglobulin domain, containing the homophilic binding site, in control of the localization of the enzyme. Deletion of the immunoglobulin domain impaired localization of PTPmu to the cell-cell contacts in endothelial and epithelial cells. In addition, deletion of the immunoglobulin domain affected the distribution of PTPmu in subconfluent endothelial cells when homophilic binding to another PTPmu molecule on an apposing cell was not possible, resulting in an accumulation of the mutant phosphatase at the cell surface with a concentration at the cell periphery in the region occupied by focal adhesions. This aberrant localization correlated with reduced survival and alterations in normal focal adhesion and cytoskeleton morphology. This study therefore illustrates the critical role of the immunoglobulin domain in regulation of the localization of PTPmu and the importance of such control for the maintenance of normal cell physiology.
Collapse
|
88
|
Lin J, Zhu JW, Baker JE, Weiss A. Regulated Expression of the Receptor-Like Tyrosine Phosphatase CD148 on Hemopoietic Cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:2324-30. [PMID: 15294945 DOI: 10.4049/jimmunol.173.4.2324] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD148 is a receptor-like protein tyrosine phosphatase expressed on a wide variety of cell types. Through the use flow cytometry and immunofluorescence microscopy on tissue sections, we examined the expression of CD148 on multiple murine hemopoietic cell lineages. We found that CD148 is moderately expressed during all stages of B cell development in the bone marrow, as well as peripheral mature B cells. In contrast, CD148 expression on thymocytes and mature T cells is substantially lower. However, stimulation of peripheral T cells through the TCR leads to an increase of CD148 expression. This up-regulation on T cells can be partially inhibited by reagents that block the activity of src family kinases, calcineurin, MEK, or PI3K. Interestingly, CD148 levels are elevated on freshly isolated T cells from MRL lpr/lpr and CTLA-4-deficient mice, two murine models of autoimmunity. Together, these expression data along with previous biochemical data suggest that CD148 may play an important regulatory role to control an immune response.
Collapse
Affiliation(s)
- Joseph Lin
- Department of Medicine, University of California, San Francisco 94143, USA
| | | | | | | |
Collapse
|
89
|
Bazzoni G, Dejana E. Endothelial cell-to-cell junctions: molecular organization and role in vascular homeostasis. Physiol Rev 2004; 84:869-901. [PMID: 15269339 DOI: 10.1152/physrev.00035.2003] [Citation(s) in RCA: 931] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Intercellular junctions mediate adhesion and communication between adjoining endothelial and epithelial cells. In the endothelium, junctional complexes comprise tight junctions, adherens junctions, and gap junctions. The expression and organization of these complexes depend on the type of vessels and the permeability requirements of perfused organs. Gap junctions are communication structures, which allow the passage of small molecular weight solutes between neighboring cells. Tight junctions serve the major functional purpose of providing a "barrier" and a "fence" within the membrane, by regulating paracellular permeability and maintaining cell polarity. Adherens junctions play an important role in contact inhibition of endothelial cell growth, paracellular permeability to circulating leukocytes and solutes. In addition, they are required for a correct organization of new vessels in angiogenesis. Extensive research in the past decade has identified several molecular components of the tight and adherens junctions, including integral membrane and intracellular proteins. These proteins interact both among themselves and with other molecules. Here, we review the individual molecules of junctions and their complex network of interactions. We also emphasize how the molecular architectures and interactions may represent a mechanistic basis for the function and regulation of junctions, focusing on junction assembly and permeability regulation. Finally, we analyze in vivo studies and highlight information that specifically relates to the role of junctions in vascular endothelial cells.
Collapse
Affiliation(s)
- Gianfranco Bazzoni
- Istituto di Ricerche Farmacologiche "Mario Negri," Via Eritrea 62, I-20157 Milan, Italy.
| | | |
Collapse
|
90
|
Persson C, Sjöblom T, Groen A, Kappert K, Engström U, Hellman U, Heldin CH, den Hertog J, Ostman A. Preferential oxidation of the second phosphatase domain of receptor-like PTP-alpha revealed by an antibody against oxidized protein tyrosine phosphatases. Proc Natl Acad Sci U S A 2004; 101:1886-91. [PMID: 14762163 PMCID: PMC357022 DOI: 10.1073/pnas.0304403101] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Protein tyrosine phosphatases (PTPs) constitute a large enzyme family with important biological functions. Inhibition of PTP activity through reversible oxidation of the active-site cysteine residue is emerging as a general, yet poorly characterized, regulatory mechanism. In this study, we describe a generic antibody-based method for detection of oxidation-inactivated PTPs. Previous observations of oxidation of receptor-like PTP (RPTP) alpha after treatment of cells with H(2)O(2) were confirmed. Platelet-derived growth factor (PDGF)-induced oxidation of endogenous SHP-2, sensitive to treatment with the phosphatidylinositol 3-kinase inhibitor LY294002, was demonstrated. Furthermore, oxidation of RPTPalpha was shown after UV-irradiation. Interestingly, the catalytically inactive second PTP domain of RPTPalpha demonstrated higher susceptibility to oxidation. The experiments thus demonstrate previously unrecognized intrinsic differences between PTP domains to susceptibility to oxidation and suggest mechanisms for regulation of RPTPs with tandem PTP domains. The antibody strategy for detection of reversible oxidation is likely to facilitate further studies on regulation of PTPs and might be applicable to analysis of redox regulation of other enzyme families with active-site cysteine residues.
Collapse
Affiliation(s)
- Camilla Persson
- Ludwig Institute for Cancer Research, Box 595, SE-751 24 Uppsala, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Kellie S, Craggs G, Bird IN, Jones GE. The tyrosine phosphatase DEP-1 induces cytoskeletal rearrangements, aberrant cell-substratum interactions and a reduction in cell proliferation. J Cell Sci 2004; 117:609-18. [PMID: 14709717 DOI: 10.1242/jcs.00879] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The receptor protein tyrosine phosphatase density-enhanced phosphatase-1 (DEP-1) has been implicated in aberrant cancer cell growth and immune cell function, however, its function within cells has yet to be properly elucidated. To investigate the cellular function of DEP-1, stable cell lines inducibly expressing DEP-1 were generated. Induction of DEP-1 expression was found to decrease PDGF-stimulated tyrosine phosphorylation of a number of cellular proteins including the PDGF receptor, and to inhibit growth factor-stimulated phosphorylation of components of the MAPK pathway, indicating that DEP-1 antagonised PDGF receptor signalling. This was supported by data showing that DEP-1 expression resulted in a reduction in cell proliferation. DEP-1-expressing cells had fewer actin-containing microfilament bundles, reduced vinculin and paxillin-containing adhesion plaques, and were defective in interactions with fibronectin. Defective cell-substratum adhesion correlated with lack of activation of FAK in DEP-1-expressing cells. Time-lapse interference reflection microscopy of live cells revealed that although small focal contacts at the leading edge were generated in DEP-1-expressing cells, they failed to mature into stable focal adhesions, as found in control cells. Further motility analysis revealed that DEP-1-expressing cells retained limited random motility, but showed no chemotaxis towards a gradient of PDGF. In addition, cell-cell contacts were disrupted, with a change in the localisation of cadherin from discrete areas of cell-cell contact to large areas of membrane interaction, and there was a parallel redistribution of beta-catenin. These results demonstrate that DEP-1 is a negative regulator of cell proliferation, cell-substratum contacts, motility and chemotaxis in fibroblasts.
Collapse
Affiliation(s)
- Stuart Kellie
- School of Molecular and Microbial Sciences, Institute for Molecular Bioscience and CRC for Chronic Inflammatory Diseases, University of Queensland, Brisbane, QLD 4072, Australia.
| | | | | | | |
Collapse
|
92
|
Ito T, Okazawa H, Maruyama K, Tomizawa K, Motegi SI, Ohnishi H, Kuwano H, Kosugi A, Matozaki T. Interaction of SAP-1, a transmembrane-type protein-tyrosine phosphatase, with the tyrosine kinase Lck. Roles in regulation of T cell function. J Biol Chem 2003; 278:34854-63. [PMID: 12837766 DOI: 10.1074/jbc.m300648200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
SAP-1 is a transmembrane-type protein-tyrosine phosphatase that is expressed in most tissues but whose physiological functions remain unknown. The cytoplasmic region of SAP-1 has now been shown to bind directly the tyrosine kinase Lck. Overexpression of wild-type SAP-1, but not that of a catalytically inactive mutant of SAP-1, inhibited both the basal and the T cell antigen receptor (TCR)-stimulated activity of Lck in human Jurkat T cell lines. Lck served as a direct substrate for dephosphorylation by SAP-1 in vitro. Overexpression of wild-type SAP-1 in Jurkat cells also: (i) inhibited both the activation of mitogen-activated protein kinase and the increase in cell surface expression of CD69 induced by TCR stimulation; (ii) reduced the extent of the TCR-induced increase in the tyrosine phosphorylation of ZAP-70 or that of LAT; (iii) reduced both the basal level of tyrosine phosphorylation of p62dok, as well as the increase in the phosphorylation of this protein induced by CD2 stimulation; and (iv) inhibited cell migration. These results thus suggest that the direct interaction of SAP-1 with Lck results in inhibition of the kinase activity of the latter and a consequent negative regulation of T cell function.
Collapse
Affiliation(s)
- Tomokazu Ito
- Biosignal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-Machi, Maebashi, Gunma 371-8512, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Abstract
Mutations in the von Hippel-Lindau (VHL) gene are involved in the VHL family cancer syndrome and sporadic renal cell carcinoma. Previous studies indicated that VHL-induced growth arrest required high cell density and growth on extracellular matrix. In the present study, VHL protein (pVHL) levels were observed to be dramatically increased in cells grown to high cell density compared to cells grown at low cell density. Reverse transcription-polymerase chain reaction and Northern blot analysis indicated that VHL mRNA levels were equivalent in sparse and dense cells. The pVHL was rapidly degraded when cell-cell contact was disturbed by trypsinization or EDTA release. Treatment of cells with a proteasome inhibitor blocked the degradation of pVHL. Using a set of VHL deletions fused to GFP, a cell density-dependent region (CDDR) was identified and localized to the c-terminus of pVHL. In addition, other members of the VBC protein complex also showed a cell density-dependent regulation similar to pVHL. Cell density regulation of VHL did not require elongin binding and density-dependent regulation of other VBC components was not dependent on pVHL. In addition, hypoxia inducible factor-2alpha protein levels were elevated in sparse cells with low levels of pVHL and reduced or absent in confluent cells containing abundant VHL. These results indicate that pVHL levels and thus function are tightly regulated by cell-cell signaling. In addition, care must be taken when interpreting studies of VHL function and subcellular localization of cells grown at subconfluent conditions.
Collapse
Affiliation(s)
- Sankar Mohan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | |
Collapse
|
94
|
Nagano H, Noguchi T, Inagaki K, Yoon S, Matozaki T, Itoh H, Kasuga M, Hayashi Y. Downregulation of stomach cancer-associated protein tyrosine phosphatase-1 (SAP-1) in advanced human hepatocellular carcinoma. Oncogene 2003; 22:4656-63. [PMID: 12879010 DOI: 10.1038/sj.onc.1206588] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
SAP-1 (stomach cancer-associated protein tyrosine phosphatase-1) is a transmembrane-type protein tyrosine phosphatase that has been implicated as a negative regulator of integrin-mediated signaling. The potential role of this enzyme in hepatocarcinogenesis has now been investigated by examining its expression in 32 surgically excised human hepatocellular carcinoma (HCC) specimens. Both immunohistochemical and immunoblot analyses revealed that normal liver tissue, as well as tissue affected by chronic hepatitis or cirrhosis, contained substantial amounts of SAP-1. The expression level of SAP-1 in 75% of well-differentiated HCCs was similar to or higher than that observed in the surrounding noncancerous tissue. In contrast, the abundance of SAP-1 in 85.7% of moderately differentiated HCCs and in all poorly differentiated HCCs was greatly reduced compared with that in the adjacent tissue. Indeed, SAP-1 was almost undetectable in 83.3% of poorly differentiated HCCs. Furthermore, expression of recombinant SAP-1 in two highly motile human HCC cell lines resulted in a change in morphology and a marked reduction in both migratory activity and growth rate. In conclusion, these results indicate that SAP-1 expression is downregulated during the dedifferentiation of human HCC, and that this downregulation may play a causal role in disease progression.
Collapse
Affiliation(s)
- Hidenobu Nagano
- Division of Diabetes, Digestive and Kidney Diseases, Department of Clinical Molecular Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Jandt E, Denner K, Kovalenko M, Ostman A, Böhmer FD. The protein-tyrosine phosphatase DEP-1 modulates growth factor-stimulated cell migration and cell-matrix adhesion. Oncogene 2003; 22:4175-85. [PMID: 12833140 DOI: 10.1038/sj.onc.1206652] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Density-enhanced protein-tyrosine phosphatase-1 (DEP-1 also CD148) is a transmembrane molecule with a single intracellular PTP domain. It has recently been proposed to function as a tumor suppressor. We have previously shown that DEP-1 dephosphorylates the activated platelet-derived growth factor (PDGF) beta-receptor in a site-selective manner (Kovalenko et al. (2000). J. Biol. Chem. 275, 16219-16226). We analysed cell lines with inducible DEP-1 expression for cellular functions of DEP-1. Several aspects of PDGFbeta-receptor signaling were negatively affected by DEP-1 expression. These include PDGF-stimulated activation of inositol trisphosphate formation, Erk1/2, p21Ras, and Src. Activation of receptor-associated phosphoinositide-3 kinase activity and of Akt/PKB were weakly attenuated at early time points of stimulation. Inhibition of PDGF-stimulated signaling depended on DEP-1 catalytic activity. Importantly, DEP-1 inhibited PDGF-stimulated cell migration. The catalytically inactive DEP-1 C1239S variant enhanced cell migration and PDGF-stimulated Erk1/2 activation, suggesting a dominant negative interference with endogenous DEP-1. In contrast to cell migration, cell-substrate adhesion was promoted by active DEP-1 and delayed or suppressed by DEP-1 C1239S, correlating with positive effects of DEP-1 on adhesion-stimulated Src kinase. We propose that negative regulation of growth-factor stimulated cell migration and promotion of cell-matrix adhesion may be related to the function of DEP-1 as tumor suppressor.
Collapse
Affiliation(s)
- Enrico Jandt
- Research Unit Molecular Cell Biology, Medical Faculty, Friedrich Schiller University, Drackendorfer str 1, D-07747 Jena, Germany
| | | | | | | | | |
Collapse
|
96
|
Young BA, Sui X, Kiser TD, Hyun SW, Wang P, Sakarya S, Angelini DJ, Schaphorst KL, Hasday JD, Cross AS, Romer LH, Passaniti A, Goldblum SE. Protein tyrosine phosphatase activity regulates endothelial cell-cell interactions, the paracellular pathway, and capillary tube stability. Am J Physiol Lung Cell Mol Physiol 2003; 285:L63-75. [PMID: 12626337 DOI: 10.1152/ajplung.00423.2002] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Protein tyrosine phosphorylation is tightly regulated through the actions of both protein tyrosine kinases and protein tyrosine phosphatases. In this study, we demonstrate that protein tyrosine phosphatase inhibition promotes tyrosine phosphorylation of endothelial cell-cell adherens junction proteins, opens an endothelial paracellular pathway, and increases both transendothelial albumin flux and neutrophil migration. Tyrosine phosphatase inhibition with sodium orthovanadate or phenylarsine oxide induced dose- and time-dependent increases in [14C]bovine serum albumin flux across postconfluent bovine pulmonary artery endothelial cell monolayers. These increases in albumin flux were coincident with actin reorganization and intercellular gap formation in both postconfluent monolayers and preformed endothelial cell capillary tubes. Vanadate (25 microM) increased tyrosine phosphorylation of endothelial cell proteins 12-fold within 1 h. Tyrosine phosphorylated proteins were immunolocalized to the intercellular boundaries, and several were identified as the endothelial cell-cell adherens junction proteins, vascular-endothelial cadherin, and beta-, gamma-, and p120-catenin as well as platelet endothelial cell adhesion molecule-1. Of note, these tyrosine phosphorylation events were not associated with disassembly of the adherens junction complex or its uncoupling from the actin cytoskeleton. The dose and time requirements for vanadate-induced increases in phosphorylation were comparable with those defined for increments in transendothelial [14C]albumin flux and neutrophil migration, and pretreatment with the tyrosine kinase inhibitor herbimycin A protected against these effects. These data suggest that protein tyrosine phosphatases and their substrates, which localize to the endothelial cell-cell boundaries, regulate adherens junctional integrity, the movement of macromolecules and cells through the endothelial paracellular pathway, and capillary tube stability.
Collapse
Affiliation(s)
- Bradford A Young
- Division of Infectious Diseases, Department of Veterans Affairs Medical Center, Baltimore 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Wheeler MA, Townsend MK, Yunker LA, Mauro LJ. Transcriptional activation of the tyrosine phosphatase gene, OST-PTP, during osteoblast differentiation. J Cell Biochem 2003; 87:363-76. [PMID: 12397596 DOI: 10.1002/jcb.10297] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Protein tyrosine phosphatases (PTPs) are critical regulators of cellular phosphorylation functioning in processes such as cell growth, differentiation, and adhesion. Osteotesticular PTP (OST) is the only characterized member of this superfamily whose expression is regulated in osteoblasts and critical for their in vitro differentiation. Such evidence would suggest that this molecule is a key modulator of signaling events during osteogenesis, yet little is known about its genetic regulation. In an effort to examine the molecular mechanisms involved in the cellular regulation of OST, we have characterized its expression in MC3T3 osteoblasts during differentiation. Northern analysis revealed that murine OST mRNA is dramatically regulated during the preosteoblast to osteoblast progression, with predominant expression in differentiated and early mineralizing osteoblasts. This expression pattern is unique to this phosphatase since, in comparison, the structurally similar receptor PTP, LAR, and the intracellular PTP1B show little change during differentiation. Cell density contributes to this upregulated expression as confluent cultures display an increase in OST transcripts within 4 h post-plating. Transient transfection of the OST promoter in differentiating MC3T3 results in a significant increase in transcriptional activation from day 0 to day 5 of differentiation, similar in timing and intensity to the observed upregulation of the endogenous gene. This activation appears to be specific to osteoblasts, since progression to a myoblast phenotype results in no change in reporter gene activity. Culturing these preosteoblast cells in the absence of critical co-factors results in an inhibition of differentiation and leads to a delayed induction of OST transcripts as well as the attenuation of transcriptional activation. These results show that the murine OST gene is regulated at the transcriptional level in an osteoblast-specific, differentiation-dependent manner during the differentiation of MC3T3 osteoblasts. Future studies will help determine the essential regulatory elements within the OST-PTP promoter and the critical signaling pathways important in this regulation.
Collapse
Affiliation(s)
- Marie A Wheeler
- Department of Animal Science-Physiology, University of Minnesota, St. Paul, Minnesota 55108, USA
| | | | | | | |
Collapse
|
98
|
Grazia Lampugnani M, Zanetti A, Corada M, Takahashi T, Balconi G, Breviario F, Orsenigo F, Cattelino A, Kemler R, Daniel TO, Dejana E. Contact inhibition of VEGF-induced proliferation requires vascular endothelial cadherin, beta-catenin, and the phosphatase DEP-1/CD148. J Cell Biol 2003; 161:793-804. [PMID: 12771128 PMCID: PMC2199373 DOI: 10.1083/jcb.200209019] [Citation(s) in RCA: 304] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Confluent endothelial cells respond poorly to the proliferative signals of VEGF. Comparing isogenic endothelial cells differing for vascular endothelial cadherin (VE-cadherin) expression only, we found that the presence of this protein attenuates VEGF-induced VEGF receptor (VEGFR) 2 phosphorylation in tyrosine, p44/p42 MAP kinase phosphorylation, and cell proliferation. VE-cadherin truncated in beta-catenin but not p120 binding domain is unable to associate with VEGFR-2 and to induce its inactivation. beta-Catenin-null endothelial cells are not contact inhibited by VE-cadherin and are still responsive to VEGF, indicating that this protein is required to restrain growth factor signaling. A dominant-negative mutant of high cell density-enhanced PTP 1 (DEP-1)//CD148 as well as reduction of its expression by RNA interference partially restore VEGFR-2 phosphorylation and MAP kinase activation. Overall the data indicate that VE-cadherin-beta-catenin complex participates in contact inhibition of VEGF signaling. Upon stimulation with VEGF, VEGFR-2 associates with the complex and concentrates at cell-cell contacts, where it may be inactivated by junctional phosphatases such as DEP-1. In sparse cells or in VE-cadherin-null cells, this phenomenon cannot occur and the receptor is fully activated by the growth factor.
Collapse
|
99
|
Harrod TR, Justement LB. Evaluating function of transmembrane protein tyrosine phosphatase CD148 in lymphocyte biology. Immunol Res 2003; 26:153-66. [PMID: 12403354 DOI: 10.1385/ir:26:1-3:153] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The transmembrane protein tyrosine phosphatase CD148 is expressed on numerous cell types, including most cells of the hematopoietic lineage. CD148 has been shown to regulate density-dependent inhibition of cell growth as well as cellular differentiation in nonhematopoietic cells and has been shown to regulate signal transduction processes in several nonlymphoid hematopoietic cell types. Analysis of CD148 expression on lymphoid cells has demonstrated that CD148 is expressed at low levels on T cells and that it is upregulated in response to activation. Several groups have observed that CD148 negatively regulates T cell activation in response to crosslinking of the T cell antigen receptor, suggesting that it may play a role in feedback inhibition of the T cell immune response. In the B cell compartment, CD 148 expression appears to be restricted to the memory subpopulation, raising the possibility that it serves a unique function in these cells, which has yet to be determined. Recent studies have shown that CD148 interacts with the PDZ domain-containing protein syntenin, raising the possibility that its function or its localization with substrates in T and B cells may be controlled through this or a related interaction with another PDZ domain protein.
Collapse
Affiliation(s)
- Thomas R Harrod
- Department of Microbiology, University of Alabama at Birmingham, 35294-3300, USA
| | | |
Collapse
|
100
|
Takahashi T, Takahashi K, St John PL, Fleming PA, Tomemori T, Watanabe T, Abrahamson DR, Drake CJ, Shirasawa T, Daniel TO. A mutant receptor tyrosine phosphatase, CD148, causes defects in vascular development. Mol Cell Biol 2003; 23:1817-31. [PMID: 12588999 PMCID: PMC151692 DOI: 10.1128/mcb.23.5.1817-1831.2003] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vascularization defects in genetic recombinant mice have defined critical roles for a number of specific receptor tyrosine kinases. Here we evaluated whether an endothelium-expressed receptor tyrosine phosphatase, CD148 (DEP-1/PTPeta), participates in developmental vascularization. A mutant allele, CD148(DeltaCyGFP), was constructed to eliminate CD148 phosphatase activity by in-frame replacement of cytoplasmic sequences with enhanced green fluorescent protein sequences. Homozygous mutant mice died at midgestation, before embryonic day 11.5 (E11.5), with vascularization failure marked by growth retardation and disorganized vascular structures. Structural abnormalities were observed as early as E8.25 in the yolk sac, prior to the appearance of intraembryonic defects. Homozygous mutant mice displayed enlarged vessels comprised of endothelial cells expressing markers of early differentiation, including VEGFR2 (Flk1), Tal1/SCL, CD31, ephrin-B2, and Tie2, with notable lack of endoglin expression. Increased endothelial cell numbers and mitotic activity indices were demonstrated. At E9.5, homozygous mutant embryos showed homogeneously enlarged primitive vessels defective in vascular remodeling and branching, with impaired pericyte investment adjacent to endothelial structures, in similarity to endoglin-deficient embryos. Developing cardiac tissues showed expanded endocardial projections accompanied by defective endocardial cushion formation. These findings implicate a member of the receptor tyrosine phosphatase family, CD148, in developmental vascular organization and provide evidence that it regulates endothelial proliferation and endothelium-pericyte interactions.
Collapse
Affiliation(s)
- Takamune Takahashi
- Nephrology Division and Center for Vascular Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|