1
|
Takahashi K, Yu A, Otsuka T, Pasic L, Narui C, He L, Ellinger P, Grundmann M, Harris RC, Takahashi T. CD148 agonistic antibody alleviates renal injury induced by chronic angiotensin II infusion in mice. BMC Nephrol 2025; 26:165. [PMID: 40165104 PMCID: PMC11959730 DOI: 10.1186/s12882-025-04070-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Angiotensin II (Ang II) plays a critical role in the progression of kidney disease. In addition to its direct signaling events, Ang II transactivates epidermal growth factor receptor (EGFR) and causes renal injury. CD148 is a transmembrane protein tyrosine phosphatase that dephosphorylates EGFR and strongly inhibits its activity. In this study, we have asked if CD148 agonistic antibody 18E1 mAb attenuates renal injury induced by chronic Ang II infusion to explore its therapeutic application. METHODS Hypertensive nephropathy was induced in mice subjected to unilateral nephrectomy (UNx) by infusing Ang II (1.4 mg/kg per day) for 6 weeks using an osmotic minipump. The 18E1 mAb or isotype control IgG were intraperitoneally injected (15 mg/kg, three times per week) to the UNx + Ang II mice for 6 weeks, and their renal phenotype was investigated. RESULTS Chronic Ang II infusion induced evident hypertension and renal injury that is indicated by elevation of plasma creatinine, urinary albumin excretion, renal hypertrophy, podocyte injury, macrophage infiltration, and the expression of alpha smooth muscle actin and collagen deposition. As compared with isotype control antibody, 18E1 mAb significantly reduced these renal changes, while it showed no effects on blood pressure. Furthermore, phospho-EGFR immunohistochemistry and immunoblotting demonstrated renal EGFR is activated in the mice that were subjected to UNx and Ang II infusion and 18E1 mAb significantly reduces EGFR phosphorylation in these kidneys as compared with isotype control treatment. CONCLUSION Agonistic CD148 antibody attenuates UNx + Ang II-induced renal injury, in part by reducing EGFR activity.
Collapse
Affiliation(s)
- Keiko Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | - Alina Yu
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | - Tadashi Otsuka
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | - Lejla Pasic
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - Chikage Narui
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | - Lilly He
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | - Philipp Ellinger
- Bayer AG Research & Development, Pharmaceuticals, 42113, Wuppertal, Germany
| | - Manuel Grundmann
- Bayer AG Research & Development, Pharmaceuticals, 42113, Wuppertal, Germany
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232, USA.
| |
Collapse
|
2
|
Bai M, Xu S, Jiang M, Guo Y, Hu D, He J, Wang T, Zhang Y, Guo Y, Zhang Y, Huang S, Jia Z, Zhang A. Meis1 Targets Protein Tyrosine Phosphatase Receptor J in Fibroblast to Retard Chronic Kidney Disease Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309754. [PMID: 39162106 PMCID: PMC11497016 DOI: 10.1002/advs.202309754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/11/2024] [Indexed: 08/21/2024]
Abstract
Renal fibrosis is a common pathological feature of chronic kidney disease (CKD) with the proliferation and activation of myofibroblasts being definite effectors and drivers. Here, increased expression of Meis1 (myeloid ecotropic viral integration site 1) is observed, predominantly in the nucleus of the kidney of CKD patients and mice, and negatively correlates with serum creatinine. Fibroblast-specific knock-in of Meis1 inhibits myofibroblast activation and attenuates renal fibrosis and kidney dysfunction in CKD models. Overexpression of Meis1 in NRK-49F cells suppresses the pro-fibrotic response induced by transforming growth factor-β1 but accelerates by its knockdown. Mechanistically, Meis1 targets protein tyrosine phosphatase receptor J (Ptprj) to block renal fibrosis by inhibiting the proliferation and activation of fibroblasts. Finally, a new activator of Ptprj is identified through computer-aided virtual screening, which has the effect of alleviating renal fibrosis. Collectively, these results illustrate that the Meis1/Ptprj axis has therapeutic potential for clinically treating CKD.
Collapse
Affiliation(s)
- Mi Bai
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Nanjing Key Lab of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008China
| | - Shuang Xu
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Mingzhu Jiang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Yuxian Guo
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Dandan Hu
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Jia He
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Ting Wang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Yu Zhang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Medical School of Nanjing UniversityNanjing210093China
| | - Yan Guo
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
| | - Yue Zhang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Nanjing Key Lab of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008China
| | - Songming Huang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Nanjing Key Lab of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008China
| | - Zhanjun Jia
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Nanjing Key Lab of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008China
| | - Aihua Zhang
- Department of Nephrology, State Key Laboratory of Reproductive MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029China
- Nanjing Key Lab of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008China
| |
Collapse
|
3
|
Bhattacharyya A, Barbee KA. Vascular endothelial cell morphology and alignment regulate VEGF-induced endothelial nitric oxide synthase activation. Cytoskeleton (Hoboken) 2024; 81:473-487. [PMID: 38775643 PMCID: PMC11496009 DOI: 10.1002/cm.21872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 06/13/2024]
Abstract
Nitric oxide (NO) production by endothelial nitric oxide synthase (eNOS) inhibits platelet and leukocyte adhesion while promoting vasorelaxation in smooth muscle cells. Dysfunctional regulation of eNOS is a hallmark of various vascular pathologies, notably atherosclerosis, often associated with areas of low shear stress on endothelial cells (ECs). While the link between EC morphology and local hemodynamics is acknowledged, the specific impact of EC morphology on eNOS regulation remains unclear. Morphological differences between elongated, aligned ECs and polygonal, randomly oriented ECs correspond to variations in focal adhesion and cytoskeletal organization, suggesting differing levels of cytoskeletal prestress. However, the functional outcomes of cytoskeletal prestress, particularly in the absence of shear stress, are not extensively studied in ECs. Some evidence suggests that elongated ECs exhibit decreased immunogenicity and enhanced NO production. This study aims to elucidate the signaling pathways governing VEGF-stimulated eNOS regulation in the aligned EC phenotype characterized by elongated and aligned cells within a monolayer. Using anisotropic topographic cues, bovine aortic endothelial cells (BAECs) were elongated and aligned, followed by VEGF treatment in the presence or absence of cytoskeletal tension inhibitors. Phosphorylation of eNOS ser1179, AKT ser437 and FAK Tyr397 in response to VEGF challenge were significantly heightened in aligned ECs compared to unaligned ECs. Moreover this response proved to be robustly tied to cytoskeletal tension as evinced by the abrogation of responses in the presence of the myosin II ATPase inhibitor, blebbistatin. Notably, this work demonstrates for the first time the reliance on FAK phosphorylation in VEGF-mediated eNOS activation and the comparatively greater contribution of the cytoskeletal machinery in propagating VEGF-eNOS signaling in aligned and elongated ECs. This research underscores the importance of utilizing appropriate vascular models in drug development and sheds light on potential mechanisms underlying vascular function and pathology that can help inform vascular graft design.
Collapse
Affiliation(s)
- Aparna Bhattacharyya
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| | - Kenneth A. Barbee
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St., Philadelphia, PA 19104, USA
| |
Collapse
|
4
|
Young KA, Wojdyla K, Lai T, Mulholland KE, Aldaz Casanova S, Antrobus R, Andrews SR, Biggins L, Mahler-Araujo B, Barton PR, Anderson KR, Fearnley GW, Sharpe HJ. The receptor protein tyrosine phosphatase PTPRK promotes intestinal repair and catalysis-independent tumour suppression. J Cell Sci 2024; 137:jcs261914. [PMID: 38904097 PMCID: PMC11298714 DOI: 10.1242/jcs.261914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
PTPRK is a receptor tyrosine phosphatase that is linked to the regulation of growth factor signalling and tumour suppression. It is stabilized at the plasma membrane by trans homophilic interactions upon cell-cell contact. PTPRK regulates cell-cell adhesion but is also reported to regulate numerous cancer-associated signalling pathways. However, the signalling mechanism of PTPRK remains to be determined. Here, we find that PTPRK regulates cell adhesion signalling, suppresses invasion and promotes collective, directed migration in colorectal cancer cells. In vivo, PTPRK supports recovery from inflammation-induced colitis. In addition, we confirm that PTPRK functions as a tumour suppressor in the mouse colon and in colorectal cancer xenografts. PTPRK regulates growth factor and adhesion signalling, and suppresses epithelial to mesenchymal transition (EMT). Contrary to the prevailing notion that PTPRK directly dephosphorylates EGFR, we find that PTPRK regulation of both EGFR and EMT is independent of its catalytic function. This suggests that additional adaptor and scaffold functions are important features of PTPRK signalling.
Collapse
Affiliation(s)
| | | | - Tiffany Lai
- Signalling programme, Babraham Institute, Cambridge CB22 3AT, UK
| | | | | | - Robin Antrobus
- Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | | | - Laura Biggins
- Bioinformatics, Babraham Institute, Cambridge CB22 3AT, UK
| | | | - Philippa R. Barton
- Cambridge Institute for Medical Research, Hills Road, Cambridge CB2 0XY, UK
| | - Keith R. Anderson
- Molecular biology department, Genentech, South San Francisco, CA 94080, USA
| | | | - Hayley J. Sharpe
- Signalling programme, Babraham Institute, Cambridge CB22 3AT, UK
| |
Collapse
|
5
|
Li MY, Gao RP, Zhu Q, Chen Y, Tao BB, Zhu YC. Skeletal muscle-derived FSTL1 starting up angiogenesis by regulating endothelial junction via activating Src pathway can be upregulated by hydrogen sulfide. Am J Physiol Cell Physiol 2023; 325:C1252-C1266. [PMID: 37694287 DOI: 10.1152/ajpcell.00219.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/12/2023]
Abstract
Hydrogen sulfide (H2S) promotes microangiogenesis and revascularization after ischemia. Neovascularization starts with the destruction of intercellular junctions and is accompanied by various endothelial cell angiogenic behaviors. Follistatin-like 1 (FSTL1) is a cardiovascular-protective myokine that works against ischemic injury. The present study examined whether FSTL1 was involved in H2S-induced angiogenesis and explored the underlying molecular mechanism. We observed that H2S accelerated blood perfusion after ischemia in the mouse hindlimb ischemia model. Western blot analysis showed that H2S stabilized FSTL1 transcript and increased FSTL1 and Human antigen R (HuR) levels in skeletal muscle. RNA-interference HuR significantly inhibited the H2S-promoted increase in FSTL1 levels. Exogenous FSTL1 promoted the wound-healing migration of human umbilical vein endothelial cells (HUVECs) and increased monolayer endothelial barrier permeability. Immunostaining showed that FSTL1 increased interendothelial gap formation and decreased VE-Cadherin, Occludin, Connexin-43, and Claudin-5 expression. In addition, FSTL1 significantly increased the phosphorylation of Src and VEGFR2. However, the Src inhibitor, not the VEGFR2 inhibitor, could block FSTL1-induced effects in angiogenesis. In conclusion, we demonstrated that H2S could upregulate the expression of FSTL1 by increasing the HuR levels in skeletal muscle, and paracrine FSTL1 could initiate angiogenesis by opening intercellular junctions via the Src signaling pathway.NEW & NOTEWORTHY The myocyte-derived paracrine protein FSTL1 acts on vascular endothelial cells and initiates the process of angiogenesis by opening the intercellular junction via activating Src kinase. H2S can significantly upregulate FSTL1 protein levels in skeletal muscles by increasing HuR expression.
Collapse
Affiliation(s)
- Meng-Yao Li
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ru-Pan Gao
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ying Chen
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Bei-Bei Tao
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yi-Chun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Chen Z, Xiao Y, Yang P, Wang R. Pan-cancer Analysis Reveals SRC May Link Lipid Metabolism and Macrophages. IRANIAN JOURNAL OF BIOTECHNOLOGY 2023; 21:e3325. [PMID: 37228626 PMCID: PMC10203188 DOI: 10.30498/ijb.2023.335402.3325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 11/09/2022] [Indexed: 05/27/2023]
Abstract
Background SRC is a member of the membrane-associated non-receptor protein tyrosine kinase superfamily. It has been reported to mediate inflammation and cancer. However, the exact molecular mechanism involved is still not clear. Objectives The current study was designed to explore the prognostic landscape of SRC and further investigate the relationship between SRC and immune infiltration in pan-cancer. Materials and Methods Kaplan-Meier Plotter was used to detect the prognostic value of SRC in pan-cancer. Then using TIMER2.0 and CIBERSORT, the relationship between SRC and immune infiltration in pan-cancer was evaluated. Furthermore, the LinkedOmics database was used to screen SRC co-expressed genes, followed by functional enrichment of SRC co-expressed genes by Metascape online tool. STRING database and Cytoscape software were applied to construct and visualise the protein-protein interaction network of SRC co-expressed genes. MCODE plug-in was used to screen hub modules in the PPI network. The SRC co-expressed genes in hub modules were extracted, and the correlation analysis between interested SRC co-expressed genes and immune infiltration was conducted via TIMER2.0 and CIBERSORT. Results Our study demonstrated that SRC expression was significantly associated with overall survival and relapse-free survival in multiple cancer types. In addition, SRC expression was significantly correlated with the immune infiltration of B cells, dendritic cells, CD4+ T cells, macrophages, and neutrophils in pan-cancer. The expression of SRC had shown to have close correlations with M1 macrophage polarisation in LIHC, TGCT, THCA, and THYM. Moreover, the genes that co-expressed with SRC in LIHC, TGCT, THCA, and THYM were mainly enriched in lipid metabolism. Besides, correlation analysis showed that SRC co-expressed genes associated with lipid metabolism were also significantly correlated with the infiltration and polarisation of macrophages. Conclusion These results indicate that SRC can serve as a prognostic biomarker in pan-cancer and is related to macrophages infiltration and interacts with genes involved in lipid metabolism.
Collapse
Affiliation(s)
- Zhongyuan Chen
- College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China
- Changde Research Centre for Artificial Intelligence and Biomedicine, Changde, Hunan, China
| | - Yaqian Xiao
- Furong College, Hunan 3Furong College, Hunan University of Arts and Science, Changde, Hunan, China
| | - Pinhong Yang
- College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China
- Changde Research Centre for Artificial Intelligence and Biomedicine, Changde, Hunan, China
| | - Ruisong Wang
- College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China
- Changde Research Centre for Artificial Intelligence and Biomedicine, Changde, Hunan, China
| |
Collapse
|
7
|
Liang X, Chen S, Wang X, Zhou L, Chen L. miR-204-5p promotes preeclampsia serum-induced injury in human umbilical vein endothelial cells through regulation of the PTPRJ/Notch axis. Pregnancy Hypertens 2022; 28:100-108. [DOI: 10.1016/j.preghy.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/28/2022]
|
8
|
He L, Takahashi K, Pasic L, Narui C, Ellinger P, Grundmann M, Takahashi T. The effects of CD148 Q276P/R326Q polymorphisms in A431D epidermoid cancer cell proliferation and epidermal growth factor receptor signaling. Cancer Rep (Hoboken) 2021; 5:e1566. [PMID: 34791835 PMCID: PMC9458507 DOI: 10.1002/cnr2.1566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/16/2021] [Accepted: 09/21/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND CD148 is a transmembrane protein tyrosine phosphatase that is expressed in multiple cell types. Previous studies have shown that CD148 dephosphorylates growth factor receptors and their signaling molecules, including EGFR and ERK1/2, and negatively regulates cancer cell growth. Furthermore, research of clinical patients has shown that highly linked CD148 gene polymorphisms, Gln276Pro (Q276P) and Arg326Gln (R326Q), are associated with an increased risk of several types of cancer. However, the biological effects of these missense mutations have not been studied. AIM We aimed to determine the biological effects of CD148 Q276P/R326Q mutations in cancer cell proliferation and growth factor signaling, with emphasis on EGFR signaling. METHODS CD148 forms, wild-type (WT) or Q276P/R326Q, were retrovirally introduced into A431D epidermoid carcinoma cells that lacks CD148 expression. The stable cells that express comparable levels of CD148 were sorted by flow cytometry. A431D cells infected with empty retrovirus was used as a control. CD148 localization, cell proliferation rate, EGFR signaling, and the response to thrombospondin-1 (TSP1), a CD148 ligand, were assessed by immunostaining, cell proliferation assay, enzyme-linked immunosorbent assay, and Western blotting. RESULTS Both CD148 forms (WT, Q276P/R326Q) were distributed to cell surface and all three cell lines expressed same level of EGFR. Compared to control cells, the A431D cells that express CD148 forms showed significantly lower cell proliferation rates. EGF-induced EGFR and ERK1/2 phosphorylation as well as cell proliferation were also significantly reduced in these cells. Furthermore, TSP1 inhibited cell proliferation in CD148 (WT, Q276P/R326Q)-expressing A431D cells, while it showed no effects in control cells. However, significant differences were not observed between CD148 WT and Q276P/R326Q cells. CONCLUSION Our data demonstrates that Q276P/R326Q mutations do not have major effects on TSP1-CD148 interaction as well as on CD148's cellular localization and activity to inhibit EGFR signaling and cell proliferation.
Collapse
Affiliation(s)
- Lilly He
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Keiko Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lejla Pasic
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Chikage Narui
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Philipp Ellinger
- Bayer AG Research & Development, Pharmaceuticals, Wuppertal, Germany
| | - Manuel Grundmann
- Bayer AG Research & Development, Pharmaceuticals, Wuppertal, Germany
| | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
9
|
Shalev M, Arman E, Stein M, Cohen-Sharir Y, Brumfeld V, Kapishnikov S, Royal I, Tuckermann J, Elson A. PTPRJ promotes osteoclast maturation and activity by inhibiting Cbl-mediated ubiquitination of NFATc1 in late osteoclastogenesis. FEBS J 2021; 288:4702-4723. [PMID: 33605542 DOI: 10.1111/febs.15778] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 01/22/2021] [Accepted: 02/18/2021] [Indexed: 11/28/2022]
Abstract
Bone-resorbing osteoclasts (OCLs) are multinucleated phagocytes, whose central roles in regulating bone formation and homeostasis are critical for normal health and development. OCLs are produced from precursor monocytes in a multistage process that includes initial differentiation, cell-cell fusion, and subsequent functional and morphological maturation; the molecular regulation of osteoclastogenesis is not fully understood. Here, we identify the receptor-type protein tyrosine phosphatase PTPRJ as an essential regulator specifically of OCL maturation. Monocytes from PTPRJ-deficient (JKO) mice differentiate and fuse normally, but their maturation into functional OCLs and their ability to degrade bone are severely inhibited. In agreement, mice lacking PTPRJ throughout their bodies or only in OCLs exhibit increased bone mass due to reduced OCL-mediated bone resorption. We further show that PTPRJ promotes OCL maturation by dephosphorylating the M-CSF receptor (M-CSFR) and Cbl, thus reducing the ubiquitination and degradation of the key osteoclastogenic transcription factor NFATc1. Loss of PTPRJ increases ubiquitination of NFATc1 and reduces its amounts at later stages of osteoclastogenesis, thereby inhibiting OCL maturation. PTPRJ thus fulfills an essential and cell-autonomous role in promoting OCL maturation by balancing between the pro- and anti-osteoclastogenic activities of the M-CSFR and maintaining NFATc1 expression during late osteoclastogenesis.
Collapse
Affiliation(s)
- Moran Shalev
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Esther Arman
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Merle Stein
- Institute of Comparative Molecular Endocrinology, University of Ulm, Germany
| | - Yael Cohen-Sharir
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Vlad Brumfeld
- Department of Chemical Research Support, The Weizmann Institute of Science, Rehovot, Israel
| | - Sergey Kapishnikov
- Department of Chemical Research Support, The Weizmann Institute of Science, Rehovot, Israel
| | - Isabelle Royal
- CRCHUM - Centre de Recherche du Centre Hospitalier de l'Université de Montréal, QC, Canada.,Institut du Cancer de Montréal, QC, Canada.,Department of Medicine, University of Montreal, QC, Canada
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Germany
| | - Ari Elson
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
10
|
Mercier C, Rousseau M, Geraldes P. Growth Factor Deregulation and Emerging Role of Phosphatases in Diabetic Peripheral Artery Disease. Front Cardiovasc Med 2021; 7:619612. [PMID: 33490120 PMCID: PMC7817696 DOI: 10.3389/fcvm.2020.619612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/10/2020] [Indexed: 01/25/2023] Open
Abstract
Peripheral artery disease is caused by atherosclerosis of lower extremity arteries leading to the loss of blood perfusion and subsequent critical ischemia. The presence of diabetes mellitus is an important risk factor that greatly increases the incidence, the progression and the severity of the disease. In addition to accelerated disease progression, diabetic patients are also more susceptible to develop serious impairment of their walking abilities through an increased risk of lower limb amputation. Hyperglycemia is known to alter the physiological development of collateral arteries in response to ischemia. Deregulation in the production of several critical pro-angiogenic factors has been reported in diabetes along with vascular cell unresponsiveness in initiating angiogenic processes. Among the multiple molecular mechanisms involved in the angiogenic response, protein tyrosine phosphatases are potent regulators by dephosphorylating pro-angiogenic tyrosine kinase receptors. However, evidence has indicated that diabetes-induced deregulation of phosphatases contributes to the progression of several micro and macrovascular complications. This review provides an overview of growth factor alterations in the context of diabetes and peripheral artery disease, as well as a description of the role of phosphatases in the regulation of angiogenic pathways followed by an analysis of the effects of hyperglycemia on the modulation of protein tyrosine phosphatase expression and activity. Knowledge of the role of phosphatases in diabetic peripheral artery disease will help the development of future therapeutics to locally regulate phosphatases and improve angiogenesis.
Collapse
Affiliation(s)
- Clément Mercier
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marina Rousseau
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pedro Geraldes
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
11
|
High expression of PTPRM predicts poor prognosis and promotes tumor growth and lymph node metastasis in cervical cancer. Cell Death Dis 2020; 11:687. [PMID: 32826853 PMCID: PMC7443137 DOI: 10.1038/s41419-020-02826-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 01/01/2023]
Abstract
The prognosis for cervical cancer (CCa) patients with lymph node metastasis (LNM) is dismal. Elucidation of the molecular mechanisms underlying LNM may provide clinical therapeutic strategies for CCa patients with LNM. However, the precise mechanism of LNM in CCa remains unclear. Herein, we demonstrated that protein tyrosine phosphatase receptor type M (PTPRM), identified from TCGA dataset, was markedly upregulated in CCa with LNM and correlated with LNM. Moreover, PTPRM was an independent prognostic factor of CCa patients in multivariate Cox′s proportional hazards model analysis and associated with poor prognosis. Furthermore, through gain-of-function and loss-of-function approaches, we found that PTPRM promoted CCa cells proliferation, migration, invasion, lymphangiogenesis, and LNM. Mechanistically, PTPRM promoted epithelial–mesenchymal transition (EMT) via Src-AKT signaling pathway and induced lymphangiogenesis in a VEGF-C dependent manner, resulting in LNM of CCa. Importantly, knockdown of PTPRM dramatically reduced LNM in vivo, suggesting that PTPRM plays an important role in the LNM of CCa. Taken together, our findings uncover a novel molecular mechanism in the LNM of CCa and identify PTPRM as a novel prognostic factor and potential therapeutic target for LNM in CCa.
Collapse
|
12
|
Song M, Finley SD. ERK and Akt exhibit distinct signaling responses following stimulation by pro-angiogenic factors. Cell Commun Signal 2020; 18:114. [PMID: 32680529 PMCID: PMC7368799 DOI: 10.1186/s12964-020-00595-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/11/2020] [Indexed: 02/07/2023] Open
Abstract
Background Angiogenesis plays an important role in the survival of tissues, as blood vessels provide oxygen and nutrients required by the resident cells. Thus, targeting angiogenesis is a prominent strategy in many different settings, including both tissue engineering and cancer treatment. However, not all of the approaches that modulate angiogenesis lead to successful outcomes. Angiogenesis-based therapies primarily target pro-angiogenic factors such as vascular endothelial growth factor-A (VEGF) or fibroblast growth factor (FGF) in isolation, and there is a limited understanding of how these promoters combine together to stimulate angiogenesis. Targeting one pathway could be insufficient, as alternative pathways may compensate, diminishing the overall effect of the treatment strategy. Methods To gain mechanistic insight and identify novel therapeutic strategies, we have developed a detailed mathematical model to quantitatively characterize the crosstalk of FGF and VEGF intracellular signaling. The model focuses on FGF- and VEGF-induced mitogen-activated protein kinase (MAPK) signaling to promote cell proliferation and the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway, which promotes cell survival and migration. We fit the model to published experimental datasets that measure phosphorylated extracellular regulated kinase (pERK) and Akt (pAkt) upon FGF or VEGF stimulation. We validate the model with separate sets of data. Results We apply the trained and validated mathematical model to characterize the dynamics of pERK and pAkt in response to the mono- and co-stimulation by FGF and VEGF. The model predicts that for certain ranges of ligand concentrations, the maximum pERK level is more responsive to changes in ligand concentration compared to the maximum pAkt level. Also, the combination of FGF and VEGF indicates a greater effect in increasing the maximum pERK compared to the summation of individual effects, which is not seen for maximum pAkt levels. In addition, our model identifies the influential species and kinetic parameters that specifically modulate the pERK and pAkt responses, which represent potential targets for angiogenesis-based therapies. Conclusions Overall, the model predicts the combination effects of FGF and VEGF stimulation on ERK and Akt quantitatively and provides a framework to mechanistically explain experimental results and guide experimental design. Thus, this model can be utilized to study the effects of pro- and anti-angiogenic therapies that particularly target ERK and/or Akt activation upon stimulation with FGF and VEGF. Video Abstract
Collapse
Affiliation(s)
- Min Song
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Stacey D Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA. .,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA. .,Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Wu Q, Finley SD. Mathematical Model Predicts Effective Strategies to Inhibit VEGF-eNOS Signaling. J Clin Med 2020; 9:jcm9051255. [PMID: 32357492 PMCID: PMC7287924 DOI: 10.3390/jcm9051255] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/12/2020] [Accepted: 04/20/2020] [Indexed: 12/27/2022] Open
Abstract
The endothelial nitric oxide synthase (eNOS) signaling pathway in endothelial cells has multiple physiological significances. It produces nitric oxide (NO), an important vasodilator, and enables a long-term proliferative response, contributing to angiogenesis. This signaling pathway is mediated by vascular endothelial growth factor (VEGF), a pro-angiogenic species that is often targeted to inhibit tumor angiogenesis. However, inhibiting VEGF-mediated eNOS signaling can lead to complications such as hypertension. Therefore, it is important to understand the dynamics of eNOS signaling in the context of angiogenesis inhibitors. Thrombospondin-1 (TSP1) is an important angiogenic inhibitor that, through interaction with its receptor CD47, has been shown to redundantly inhibit eNOS signaling. However, the exact mechanisms of TSP1's inhibitory effects on this pathway remain unclear. To address this knowledge gap, we established a molecular-detailed mechanistic model to describe VEGF-mediated eNOS signaling, and we used the model to identify the potential intracellular targets of TSP1. In addition, we applied the predictive model to investigate the effects of several approaches to selectively target eNOS signaling in cells experiencing high VEGF levels present in the tumor microenvironment. This work generates insights for pharmacologic targets and therapeutic strategies to inhibit tumor angiogenesis signaling while avoiding potential side effects in normal vasoregulation.
Collapse
Affiliation(s)
- Qianhui Wu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA;
| | - Stacey D. Finley
- Department of Biomedical Engineering, Mork Family Department of Chemical Engineering and Materials Science, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence: ; Tel.: +1-213-740-8788
| |
Collapse
|
14
|
Viallard C, Audiger C, Popovic N, Akla N, Lanthier K, Legault-Navarrete I, Melichar H, Costantino S, Lesage S, Larrivée B. BMP9 signaling promotes the normalization of tumor blood vessels. Oncogene 2020; 39:2996-3014. [PMID: 32042114 DOI: 10.1038/s41388-020-1200-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/10/2020] [Accepted: 01/29/2020] [Indexed: 01/09/2023]
Abstract
The presence of an immature tumor vascular network contributes to cancer dissemination and the development of resistance to therapies. Strategies to normalize the tumor vasculature are therefore of significant therapeutic interest for cancer treatments. VEGF inhibitors are used clinically to normalize tumor blood vessels. However, the time frame and dosage of these inhibitors required to achieve normalization is rather narrow, and there is a need to identify additional signaling targets to attain vascular normalization. In addition to VEGF, the endothelial-specific receptor Alk1 plays a critical role in vascular development and promotes vascular remodeling and maturation. Therefore, we sought to evaluate the effects of the Alk1 ligand BMP9 on tumor vascular formation. BMP9 overexpression in Lewis Lung Carcinoma (LLC) tumors significantly delayed tumor growth. Blood vessels in BMP9-overexpressing LLC tumors displayed markers of vascular maturation and were characterized by increased perivascular cell coverage. Tumor vasculature normalization was associated with decreased permeability and increased perfusion. These changes in vascular function in BMP9-overexpressing LLC tumors resulted in significant alterations of the tumor microenvironment, characterized by a decrease in hypoxia and an increase in immune infiltration. In conclusion, we show that BMP9 promotes vascular normalization in LLC tumors that leads to changes in the microenvironment.
Collapse
Affiliation(s)
- Claire Viallard
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Biologie Moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Cindy Audiger
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Natalija Popovic
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Biologie Moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Naoufal Akla
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Biochimie, Université de Montréal, Montréal, QC, Canada
| | - Kevin Lanthier
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Biologie Moléculaire, Université de Montréal, Montréal, QC, Canada
| | | | - Heather Melichar
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Santiago Costantino
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département d'Ophtalmologie, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Lesage
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Bruno Larrivée
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada. .,Département de Biologie Moléculaire, Université de Montréal, Montréal, QC, Canada. .,Département d'Ophtalmologie, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
15
|
Takahashi K, Kim RH, Pasic L, He L, Nagasaka S, Katagiri D, May T, Shimizu A, Harris RC, Mernaugh RL, Takahashi T. Agonistic anti-CD148 monoclonal antibody attenuates diabetic nephropathy in mice. Am J Physiol Renal Physiol 2020; 318:F647-F659. [PMID: 31984788 DOI: 10.1152/ajprenal.00288.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
CD148 is a transmembrane protein tyrosine phosphatase (PTP) that is expressed in the renal vasculature, including the glomerulus. Previous studies have shown that CD148 plays a role in the negative regulation of growth factor signals (including epidermal growth factor and vascular endothelial growth factor), suppressing cell proliferation and transformation. However, the role of CD148 in kidney disease remains unknown. Here, we generated an agonistic anti-CD148 antibody and evaluated its effects in murine diabetic nephropathy (DN). Monoclonal antibodies (mAbs) against the mouse CD148 ectodomain sequence were generated by immunizing CD148 knockout (CD148KO) mice. The mAbs that increased CD148 activity were selected by biological (proliferation) and biochemical (PTP activity) assays. The mAb (18E1) that showed strong agonistic activity was injected (10 mg/kg ip) in streptozotocin-induced wild-type and CD148KO diabetic mice for 6 wk, and the renal phenotype was then assessed. The effects of 18E1 mAb in podocyte growth factor signals were also assessed in culture. Compared with control IgG, 18E1 mAb significantly decreased albuminuria and mesangial expansion without altering hyperglycemia and blood pressure in wild-type diabetic mice. Immunohistochemical evaluation showed that 18E1 mAb significantly prevented the reduction of podocyte number and nephrin expression and decreased glomerular fibronectin expression and renal macrophage infiltration. The 18E1 mAb showed no effects in CD148KO diabetic mice. Furthermore, we demonstrated that 18E1 mAb reduces podocyte epidermal growth factor receptor signals in culture and in diabetic mice. These findings suggest that agonistic anti-CD148 mAb attenuates DN in mice, in part by reducing epidermal growth factor receptor signals in podocytes. This antibody may be used for the treatment of early DN.
Collapse
Affiliation(s)
- Keiko Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rachel H Kim
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lejla Pasic
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
| | - Lilly He
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shinya Nagasaka
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Daisuke Katagiri
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tracy May
- Vanderbilt University Antibody and Protein Resource, Nashville, Tennessee
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
16
|
Huang SW, Yang HY, Huang WJ, Chen WC, Yu MC, Wang SW, Hsu YF, Hsu MJ. WMJ-S-001, a Novel Aliphatic Hydroxamate-Based Compound, Suppresses Lymphangiogenesis Through p38mapk-p53-survivin Signaling Cascade. Front Oncol 2019; 9:1188. [PMID: 31781495 PMCID: PMC6851263 DOI: 10.3389/fonc.2019.01188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
Background and purpose: Angiogenesis and lymphangiogenesis are major routes for metastatic spread of tumor cells. It thus represent the rational targets for therapeutic intervention of cancer. Recently, we showed that a novel aliphatic hydroxamate-based compound, WMJ-S-001, exhibits anti-angiogenic, anti-inflammatory and anti-tumor properties. However, whether WMJ-S-001 is capable of suppressing lymphangiogenesis remains unclear. We are thus interested in exploring WMJ-S-001's anti-lymphangiogenic mechanisms in lymphatic endothelial cell (LECs). Experimental approach: WMJ-S-001's effects on LEC proliferation, migration and invasion, as well as signaling molecules activation were analyzed by immunoblotting, flow-cytometry, MTT, BrdU, migration and invasion assays. We performed tube formation assay to examine WMJ-S-001's ex vivo anti-lymphangiogenic effects. Key results: WMJ-S-001 inhibited serum-induced cell proliferation, migration, invasion in murine LECs (SV-LECs). WMJ-S-001 reduced the mRNA and protein levels of survivin. Survivin siRNA significantly suppressed serum-induced SV-LEC invasion. WMJ-S-001 induced p53 phosphorylation and increased its reporter activities. In addition, WMJ-S-001 increased p53 binding to the promoter region of survivin, while Sp1 binding to the region was decreased. WMJ-S-001 induced p38 mitogen-activated protein kinase (p38MAPK) activation. p38MPAK signaling blockade significantly inhibited p53 phosphorylation and restored survivin reduction in WMJ-S-001-stimulated SV-LCEs. Furthermore, WMJ-S-001 induced survivin reduction and inhibited cell proliferation, invasion and tube formation of primary human LECs. Conclusions and Implications: These observations indicate that WMJ-S-001 may suppress lymphatic endothelial remodeling and reduce lymphangiogenesis through p38MAPK-p53-survivin signaling. It also suggests that WMJ-S-001 is a potential lead compound in developing novel agents for the treatment of lymphangiogenesis-associated diseases and cancer.
Collapse
Affiliation(s)
- Shiu-Wen Huang
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yu Yang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Jan Huang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chuan Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Meng-Chieh Yu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ya-Fen Hsu
- Division of General Surgery, Department of Surgery, Landseed Hospital, Taoyuan, Taiwan
| | - Ming-Jen Hsu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
17
|
Bloch E, Sikorski EL, Pontoriero D, Day EK, Berger BW, Lazzara MJ, Thévenin D. Disrupting the transmembrane domain-mediated oligomerization of protein tyrosine phosphatase receptor J inhibits EGFR-driven cancer cell phenotypes. J Biol Chem 2019; 294:18796-18806. [PMID: 31676686 DOI: 10.1074/jbc.ra119.010229] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/21/2019] [Indexed: 12/19/2022] Open
Abstract
Receptor protein tyrosine phosphatases (RPTPs) play critical regulatory roles in mammalian signal transduction. However, the structural basis for the regulation of their catalytic activity is not fully understood, and RPTPs are generally not therapeutically targetable. This knowledge gap is partially due to the lack of known natural ligands or selective agonists of RPTPs. Contrary to what is known from structure-function studies of receptor tyrosine kinases (RTKs), RPTP activities have been reported to be suppressed by dimerization, which may prevent RPTPs from accessing their RTK substrates. We report here that homodimerization of protein tyrosine phosphatase receptor J (PTPRJ, also known as DEP-1) is regulated by specific transmembrane (TM) residues. We found that disrupting these interactions destabilizes homodimerization of full-length PTPRJ in cells, reduces the phosphorylation of the known PTPRJ substrate epidermal growth factor receptor (EGFR) and of other downstream signaling effectors, antagonizes EGFR-driven cell phenotypes, and promotes substrate access. We demonstrate these observations in human cancer cells using mutational studies and identified a peptide that binds to the PTPRJ TM domain and represents the first example of an allosteric agonist of RPTPs. The results of our study provide fundamental structural and functional insights into how PTPRJ activity is tuned by TM interactions in cells. Our findings also open up opportunities for developing peptide-based agents that could be used as tools to probe RPTPs' signaling mechanisms or to manage cancers driven by RTK signaling.
Collapse
Affiliation(s)
- Elizabeth Bloch
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015
| | - Eden L Sikorski
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015
| | - David Pontoriero
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Evan K Day
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Bryan W Berger
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Matthew J Lazzara
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Damien Thévenin
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015.
| |
Collapse
|
18
|
The protein tyrosine phosphatase PTPRJ/DEP-1 contributes to the regulation of the Notch-signaling pathway and sprouting angiogenesis. Angiogenesis 2019; 23:145-157. [DOI: 10.1007/s10456-019-09683-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022]
|
19
|
Song M, Finley SD. Mechanistic insight into activation of MAPK signaling by pro-angiogenic factors. BMC SYSTEMS BIOLOGY 2018; 12:145. [PMID: 30591051 PMCID: PMC6307205 DOI: 10.1186/s12918-018-0668-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/30/2018] [Indexed: 01/14/2023]
Abstract
Background Angiogenesis is important in physiological and pathological conditions, as blood vessels provide nutrients and oxygen needed for tissue growth and survival. Therefore, targeting angiogenesis is a prominent strategy in both tissue engineering and cancer treatment. However, not all of the approaches to promote or inhibit angiogenesis lead to successful outcomes. Angiogenesis-based therapies primarily target pro-angiogenic factors such as vascular endothelial growth factor-A (VEGF) or fibroblast growth factor (FGF) in isolation. However, pre-clinical and clinical evidence shows these therapies often have limited effects. To improve therapeutic strategies, including targeting FGF and VEGF in combination, we need a quantitative understanding of the how the promoters combine to stimulate angiogenesis. Results In this study, we trained and validated a detailed mathematical model to quantitatively characterize the crosstalk of FGF and VEGF intracellular signaling. This signaling is initiated by FGF binding to the FGF receptor 1 (FGFR1) and heparan sulfate glycosaminoglycans (HSGAGs) or VEGF binding to VEGF receptor 2 (VEGFR2) to promote downstream signaling. The model focuses on FGF- and VEGF-induced mitogen-activated protein kinase (MAPK) signaling and phosphorylation of extracellular regulated kinase (ERK), which promotes cell proliferation. We apply the model to predict the dynamics of phosphorylated ERK (pERK) in response to the stimulation by FGF and VEGF individually and in combination. The model predicts that FGF and VEGF have differential effects on pERK. Additionally, since VEGFR2 upregulation has been observed in pathological conditions, we apply the model to investigate the effects of VEGFR2 density and trafficking parameters. The model predictions show that these parameters significantly influence the response to VEGF stimulation. Conclusions The model agrees with experimental data and is a framework to synthesize and quantitatively explain experimental studies. Ultimately, the model provides mechanistic insight into FGF and VEGF interactions needed to identify potential targets for pro- or anti-angiogenic therapies. Electronic supplementary material The online version of this article (10.1186/s12918-018-0668-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min Song
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Stacey D Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA. .,Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA. .,Department of Biological Sciences, Computational Biology section, University of Southern California, 1042 Downey Way, CRB 140, Los Angeles, CA, 90089, USA.
| |
Collapse
|
20
|
Hendricks WPD, Zismann V, Sivaprakasam K, Legendre C, Poorman K, Tembe W, Perdigones N, Kiefer J, Liang W, DeLuca V, Stark M, Ruhe A, Froman R, Duesbery NS, Washington M, Aldrich J, Neff MW, Huentelman MJ, Hayward N, Brown K, Thamm D, Post G, Khanna C, Davis B, Breen M, Sekulic A, Trent JM. Somatic inactivating PTPRJ mutations and dysregulated pathways identified in canine malignant melanoma by integrated comparative genomic analysis. PLoS Genet 2018; 14:e1007589. [PMID: 30188888 PMCID: PMC6126841 DOI: 10.1371/journal.pgen.1007589] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/24/2018] [Indexed: 01/11/2023] Open
Abstract
Canine malignant melanoma, a significant cause of mortality in domestic dogs, is a powerful comparative model for human melanoma, but little is known about its genetic etiology. We mapped the genomic landscape of canine melanoma through multi-platform analysis of 37 tumors (31 mucosal, 3 acral, 2 cutaneous, and 1 uveal) and 17 matching constitutional samples including long- and short-insert whole genome sequencing, RNA sequencing, array comparative genomic hybridization, single nucleotide polymorphism array, and targeted Sanger sequencing analyses. We identified novel predominantly truncating mutations in the putative tumor suppressor gene PTPRJ in 19% of cases. No BRAF mutations were detected, but activating RAS mutations (24% of cases) occurred in conserved hotspots in all cutaneous and acral and 13% of mucosal subtypes. MDM2 amplifications (24%) and TP53 mutations (19%) were mutually exclusive. Additional low-frequency recurrent alterations were observed amidst low point mutation rates, an absence of ultraviolet light mutational signatures, and an abundance of copy number and structural alterations. Mutations that modulate cell proliferation and cell cycle control were common and highlight therapeutic axes such as MEK and MDM2 inhibition. This mutational landscape resembles that seen in BRAF wild-type and sun-shielded human melanoma subtypes. Overall, these data inform biological comparisons between canine and human melanoma while suggesting actionable targets in both species.
Collapse
Affiliation(s)
- William P. D. Hendricks
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Victoria Zismann
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Karthigayini Sivaprakasam
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Department of Biomedical Informatics, Arizona State University, Phoenix, Arizona, United States of America
| | - Christophe Legendre
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Kelsey Poorman
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States of America
- Department of Dermatology, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Waibhav Tembe
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Nieves Perdigones
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Jeffrey Kiefer
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Winnie Liang
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Valerie DeLuca
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- School of Life Sciences, Arizona State University, Phoenix, Arizona, United States of America
| | - Mitchell Stark
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Alison Ruhe
- Veterinary Genetics Laboratory, University of California Davis, Davis, California, United States of America
| | - Roe Froman
- Laboratory of Cancer and Developmental Cell Biology, Van Andel Research Institute (VARI), Grand Rapids, Michigan, United States of America
| | | | - Megan Washington
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Jessica Aldrich
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Mark W. Neff
- Program in Canine Genetics and Genomics, Van Andel Research Institute (VARI), Grand Rapids, Michigan, United States of America
| | - Matthew J. Huentelman
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Nicholas Hayward
- Oncogenomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Kevin Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Douglas Thamm
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, United States of America
| | - Gerald Post
- The Veterinary Cancer Center, Norwalk, Connecticut, United States of America
| | - Chand Khanna
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Barbara Davis
- Innogenics Inc., Harvard, Massachusetts, United States of America
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States of America
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States of America
| | - Alexander Sekulic
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Department of Dermatology, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Jeffrey M. Trent
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| |
Collapse
|
21
|
Bazzazi H, Zhang Y, Jafarnejad M, Isenberg JS, Annex BH, Popel AS. Computer Simulation of TSP1 Inhibition of VEGF-Akt-eNOS: An Angiogenesis Triple Threat. Front Physiol 2018; 9:644. [PMID: 29899706 PMCID: PMC5988849 DOI: 10.3389/fphys.2018.00644] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/11/2018] [Indexed: 01/08/2023] Open
Abstract
The matricellular protein thrombospondin-1 (TSP1) is a potent inhibitor of angiogenesis. Specifically, TSP1 has been experimentally shown to inhibit signaling downstream of vascular endothelial growth factor (VEGF). The molecular mechanism of this inhibition is not entirely clear. We developed a detailed computational model of VEGF signaling to Akt-endothelial nitric oxide synthase (eNOS) to investigate the quantitative molecular mechanism of TSP1 inhibition. The model demonstrated that TSP1 acceleration of VEGFR2 degradation is sufficient to explain the inhibition of VEGFR2 and eNOS phosphorylation. However, Akt inhibition requires TSP1-induced phosphatase recruitment to VEGFR2. The model was then utilized to test various strategies for the rescue of VEGF signaling to Akt and eNOS. Inhibiting TSP1 was predicted to be not as effective as CD47 depletion in rescuing signaling to Akt. The model further predicts that combination strategy involving depletion of CD47 and inhibition of TSP1 binding to CD47 is necessary for effective recovery of signaling to eNOS. In all, computational modeling offers insight to molecular mechanisms involving TSP1 interaction with VEGF signaling and provides strategies for rescuing angiogenesis by targeting TSP1-CD47 axis.
Collapse
Affiliation(s)
- Hojjat Bazzazi
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Jeffrey S Isenberg
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States.,Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brian H Annex
- Division of Cardiovascular Medicine, Department of Medicine, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
22
|
Bilotta A, Dattilo V, D'Agostino S, Belviso S, Scalise S, Bilotta M, Gaudio E, Paduano F, Perrotti N, Florio T, Fusco A, Iuliano R, Trapasso F. A novel splice variant of the protein tyrosine phosphatase PTPRJ that encodes for a soluble protein involved in angiogenesis. Oncotarget 2018; 8:10091-10102. [PMID: 28052032 PMCID: PMC5354644 DOI: 10.18632/oncotarget.14350] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/13/2016] [Indexed: 02/01/2023] Open
Abstract
PTPRJ is a receptor protein tyrosine phosphatase with tumor suppressor activity. Very little is known about the role of PTPRJ ectodomain, although recently both physiological and synthetic PTPRJ ligands have been identified. A putative shorter spliced variant, coding for a 539 aa protein corresponding to the extracellular N-terminus of PTPRJ, is reported in several databases but, currently, no further information is available. Here, we confirmed that the PTPRJ short isoform (named sPTPRJ) is a soluble protein secreted into the supernatant of both endothelial and tumor cells. Like PTPRJ, also sPTPRJ undergoes post-translational modifications such as glycosylation, as assessed by sPTPRJ immunoprecipitation. To characterize its functional activity, we performed an endothelial cell tube formation assay and a wound healing assay on HUVEC cells overexpressing sPTPRJ and we found that sPTPRJ has a proangiogenic activity. We also showed that sPTPRJ expression down-regulates endothelial adhesion molecules, that is a hallmark of proangiogenic activity. Moreover, sPTPRJ mRNA levels in human high-grade glioma, one of the most angiogenic tumors, are higher in tumor samples compared to controls. Further studies will be helpful not only to clarify the way sPTPRJ works but also to supply clues to circumvent its activity in cancer therapy.
Collapse
Affiliation(s)
- Anna Bilotta
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Vincenzo Dattilo
- Department of Scienze della Salute, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Sabrina D'Agostino
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Stefania Belviso
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Stefania Scalise
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Mariaconcetta Bilotta
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Eugenio Gaudio
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy.,Lymphoma and Genomics Research Program, Institute of Oncology Research (IOR), Bellinzona, Switzerland
| | - Francesco Paduano
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy.,Tecnologica Research Institute, Biomedical Section, Crotone, Italy
| | - Nicola Perrotti
- Department of Scienze della Salute, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Tullio Florio
- Laboratory of Pharmacology, Dept. of Internal Medicine, and Center of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy
| | - Alfredo Fusco
- Istituto di Endocrinologia e Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University Federico II of Napoli, Napoli, Italy
| | - Rodolfo Iuliano
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Francesco Trapasso
- Department of Medicina Sperimentale e Clinica, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
23
|
Regulation of receptor-type protein tyrosine phosphatases by their C-terminal tail domains. Biochem Soc Trans 2017; 44:1295-1303. [PMID: 27911712 DOI: 10.1042/bst20160141] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/06/2016] [Accepted: 07/11/2016] [Indexed: 01/10/2023]
Abstract
Protein tyrosine phosphatases (PTPs) perform specific functions in vivo, despite being vastly outnumbered by their substrates. Because of this and due to the central roles PTPs play in regulating cellular function, PTP activity is regulated by a large variety of molecular mechanisms. We review evidence that indicates that the divergent C-terminal tail sequences (C-terminal domains, CTDs) of receptor-type PTPs (RPTPs) help regulate RPTP function by controlling intermolecular associations in a way that is itself subject to physiological regulation. We propose that the CTD of each RPTP defines an 'interaction code' that helps determine molecules it will interact with under various physiological conditions, thus helping to regulate and diversify PTP function.
Collapse
|
24
|
Computational investigation of sphingosine kinase 1 (SphK1) and calcium dependent ERK1/2 activation downstream of VEGFR2 in endothelial cells. PLoS Comput Biol 2017; 13:e1005332. [PMID: 28178265 PMCID: PMC5298229 DOI: 10.1371/journal.pcbi.1005332] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/23/2016] [Indexed: 01/14/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a powerful regulator of neovascularization. VEGF binding to its cognate receptor, VEGFR2, activates a number of signaling pathways including ERK1/2. Activation of ERK1/2 is experimentally shown to involve sphingosine kinase 1 (SphK1) activation and its calcium-dependent translocation downstream of ERK1/2. Here we construct a rule-based computational model of signaling downstream of VEGFR2, by including SphK1 and calcium positive feedback mechanisms, and investigate their consequences on ERK1/2 activation. The model predicts the existence of VEGF threshold in ERK1/2 activation that can be continuously tuned by cellular concentrations of SphK1 and sphingosine 1 phosphate (S1P). The computer model also predicts powerful effects of perturbations in plasma and ER calcium pump rates and the current through the CRAC channels on ERK1/2 activation dynamics, highlighting the critical role of intracellular calcium in shaping the pERK1/2 signal. The model is then utilized to simulate anti-angiogenic therapeutic interventions targeting VEGFR2-ERK1/2 axis. Simulations indicate that monotherapies that exclusively target VEGFR2 phosphorylation, VEGF, or VEGFR2 are ineffective in shutting down signaling to ERK1/2. By simulating therapeutic strategies that target multiple nodes of the pathway such as Raf and SphK1, we conclude that combination therapy should be much more effective in blocking VEGF signaling to EKR1/2. The model has important implications for interventions that target signaling pathways in angiogenesis relevant to cancer, vascular diseases, and wound healing. Vascular endothelial growth factor (VEGF) signaling is a potent regulator of angiogenesis, the growth and development of new vessels out of a preexisting vascular network. Angiogenesis requires enhanced survival, proliferation, and motility of the vascular endothelial cells. Crucial signaling endpoints in VEGF-mediated angiogenic response include elevation in intracellular calcium and the activation of the proteins ERK1 and 2 (ERK1/2). In this study, we have developed a novel computer model for the activation of ERK1/2 and calcium downstream of VEGF receptor type 2 (VEGFR2). Our model is the first of its kind to incorporate and investigate the consequences of calcium elevation and the role of a cellular lipid modifier known as sphingosine kinase 1 (SphK1). We also utilize the model to simulate therapeutic strategies targeting VEGF signaling to ERK1/2 indicating inefficiency of single therapies known as tyrosine kinase inhibitors (TKI) that target receptor phosphorylation. Computer simulations indicate that combination therapy is essential for effective blockade of this important pathway. Our results have important implications for human diseases such as cancer where plethora of anti-VEGF therapies are currently employed. Overall, our computer model sheds new light on a complex feedback involving SphK1 and calcium that radically alters the response of cells to VEGF.
Collapse
|
25
|
Bazzazi H, Isenberg JS, Popel AS. Inhibition of VEGFR2 Activation and Its Downstream Signaling to ERK1/2 and Calcium by Thrombospondin-1 (TSP1): In silico Investigation. Front Physiol 2017; 8:48. [PMID: 28220078 PMCID: PMC5292565 DOI: 10.3389/fphys.2017.00048] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/17/2017] [Indexed: 12/13/2022] Open
Abstract
VEGF signaling through VEGFR2 is a central regulator of the angiogenic response. Inhibition of VEGF signaling by the stress-induced matricellular protein TSP1 plays a role in modulating the angiogenic response to VEGF in both health and disease. TSP1 binding to CD47 inhibits VEGFR2 activation. The full implications of this inhibitory interaction are unknown. We developed a detailed rule-based computational model to inquire if TSP1-CD47 signaling through VEGF had downstream effects upon ERK1/2 and calcium. Our Simulations suggest that enhanced degradation of VEGFR2 initiated by the binding of TSP1 to CD47 is sufficient to explain the inhibition of VEGFR2 phosphorylation, calcium elevation, and ERK1/2 activation downstream of VEGF. A complementary mechanism involving the recruitment of phosphatases to the VEGFR2 complex with consequent increase in the rate of receptor dephosphorylation may augment the inhibition of the VEGF signal. The model was then utilized to simulate the effect of inhibiting external TSP1 or the depletion of CD47 as potential therapeutic strategies in restoring VEGF signaling. Results suggest that depleting CD47 is a more efficient strategy in inhibiting the effects of TSP1/CD47 on VEGF signaling. Our results highlight the utility of in silico investigations in elucidating and clarifying molecular mechanisms at the intersection of TSP1 and VEGF biology and in differentiating between competing pro-angiogenic therapeutic strategies relevant to peripheral arterial disease (PAD) and wound healing.
Collapse
Affiliation(s)
- Hojjat Bazzazi
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| | - Jeffery S Isenberg
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh Pittsburgh, PA, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
26
|
Corti F, Simons M. Modulation of VEGF receptor 2 signaling by protein phosphatases. Pharmacol Res 2017; 115:107-123. [PMID: 27888154 PMCID: PMC5205541 DOI: 10.1016/j.phrs.2016.11.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022]
Abstract
Phosphorylation of serines, threonines, and tyrosines is a central event in signal transduction cascades in eukaryotic cells. The phosphorylation state of any particular protein reflects a balance of activity between kinases and phosphatases. Kinase biology has been exhaustively studied and is reasonably well understood, however, much less is known about phosphatases. A large body of evidence now shows that protein phosphatases do not behave as indiscriminate signal terminators, but can function both as negative or positive regulators of specific signaling pathways. Genetic models have also shown that different protein phosphatases play precise biological roles in health and disease. Finally, genome sequencing has unveiled the existence of many protein phosphatases and associated regulatory subunits comparable in number to kinases. A wide variety of roles for protein phosphatase roles have been recently described in the context of cancer, diabetes, hereditary disorders and other diseases. In particular, there have been several recent advances in our understanding of phosphatases involved in regulation of vascular endothelial growth factor receptor 2 (VEGFR2) signaling. The receptor is the principal signaling molecule mediating a wide spectrum of VEGF signal and, thus, is of paramount significance in a wide variety of diseases ranging from cancer to cardiovascular to ophthalmic. This review focuses on the current knowledge about protein phosphatases' regulation of VEGFR2 signaling and how these enzymes can modulate its biological effects.
Collapse
Affiliation(s)
- Federico Corti
- Yale Cardiovascular Research Center, Department of Internal Medicine and Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine and Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
27
|
Fournier P, Dussault S, Fusco A, Rivard A, Royal I. Tyrosine Phosphatase PTPRJ/DEP-1 Is an Essential Promoter of Vascular Permeability, Angiogenesis, and Tumor Progression. Cancer Res 2016; 76:5080-91. [PMID: 27364551 DOI: 10.1158/0008-5472.can-16-1071] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/24/2016] [Indexed: 11/16/2022]
Abstract
The protein tyrosine phosphatase PTPRJ/DEP-1 has been implicated in negative growth regulation in endothelial cells, where its expression varies at transitions between proliferation and contact inhibition. However, in the same cells, DEP-1 has also been implicated in VEGF-dependent Src activation, permeability, and capillary formation, suggesting a positive role in regulating these functions. To resolve this dichotomy in vivo, we investigated postnatal angiogenesis and vascular permeability in a DEP-1-deficient mouse. In this study, we report that DEP-1 is required for Src activation and phosphorylation of its endothelial cell-specific substrate, VE-cadherin, after systemic injection of VEGF. Accordingly, VEGF-induced vascular leakage was abrogated in the DEP-1-deficient mice. Furthermore, capillary formation was impaired in murine aortic tissue rings or Matrigel plugs infused with VEGF. In the absence of DEP-1, angiogenesis triggered by ischemia or during tumor formation was defective, which in the latter case was associated with reduced tumor cell proliferation and increased apoptosis. Macrophage infiltration was also impaired, reflecting reduced vascular permeability in the tumors or a possible cell autonomous effect of DEP-1. Consequently, the formation of spontaneous and experimental lung metastases was strongly decreased in DEP-1-deficient mice. In clinical specimens of cancer, less vascularized tumors exhibited lower microvascular expression of DEP-1. Altogether, our results established DEP-1 as an essential driver of VEGF-dependent permeability, angiogenesis, and metastasis, suggesting a novel therapeutic route to cancer treatment. Cancer Res; 76(17); 5080-91. ©2016 AACR.
Collapse
Affiliation(s)
- Patrick Fournier
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada. Institut du cancer de Montréal, Montréal, Quebec, Canada
| | - Sylvie Dussault
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada
| | - Alfredo Fusco
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Alain Rivard
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada. Département de Médecine, Université de Montréal, Montréal, Quebec, Canada
| | - Isabelle Royal
- CRCHUM - Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada. Institut du cancer de Montréal, Montréal, Quebec, Canada. Département de Médecine, Université de Montréal, Montréal, Quebec, Canada.
| |
Collapse
|
28
|
Takahashi K, Sumarriva K, Kim R, Jiang R, Brantley-Sieders DM, Chen J, Mernaugh RL, Takahashi T. Determination of the CD148-Interacting Region in Thrombospondin-1. PLoS One 2016; 11:e0154916. [PMID: 27149518 PMCID: PMC4858292 DOI: 10.1371/journal.pone.0154916] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 04/21/2016] [Indexed: 11/28/2022] Open
Abstract
CD148 is a transmembrane protein tyrosine phosphatase that is expressed in multiple cell types, including vascular endothelial cells and duct epithelial cells. Previous studies have shown a prominent role of CD148 to reduce growth factor signals and suppress cell proliferation and transformation. Further, we have recently shown that thrombospondin-1 (TSP1) serves as a functionally important ligand for CD148. TSP1 has multiple structural elements and interacts with various cell surface receptors that exhibit differing effects. In order to create the CD148-specific TSP1 fragment, here we investigated the CD148-interacting region in TSP1 using a series of TSP1 fragments and biochemical and biological assays. Our results demonstrate that: 1) CD148 binds to the 1st type 1 repeat in TSP1; 2) Trimeric TSP1 fragments that contain the 1st type repeat inhibit cell proliferation in A431D cells that stably express wild-type CD148 (A431D/CD148wt cells), while they show no effects in A431D cells that lack CD148 or express a catalytically inactive form of CD148. The anti-proliferative effect of the TSP1 fragment in A431D/CD148wt cells was largely abolished by CD148 knockdown and antagonized by the 1st, but not the 2nd and 3rd, type 1 repeat fragment. Furthermore, the trimeric TSP1 fragments containing the 1st type repeat increased the catalytic activity of CD148 and reduced phospho-tyrosine contents of EGFR and ERK1/2, defined CD148 substrates. These effects were not observed in the TSP1 fragments that lack the 1st type 1 repeat. Last, we demonstrate that the trimeric TSP1 fragment containing the 1st type 1 repeat inhibits endothelial cell proliferation in culture and angiogenesis in vivo. These effects were largely abolished by CD148 knockdown or deficiency. Collectively, these findings indicate that the 1st type 1 repeat interacts with CD148, reducing growth factor signals and inhibiting epithelial or endothelial cell proliferation and angiogenesis.
Collapse
Affiliation(s)
- Keiko Takahashi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Katherine Sumarriva
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Rachel Kim
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Rosie Jiang
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Dana M. Brantley-Sieders
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Jin Chen
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Raymond L. Mernaugh
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Takamune Takahashi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| |
Collapse
|
29
|
Regulation of Endothelial Adherens Junctions by Tyrosine Phosphorylation. Mediators Inflamm 2015; 2015:272858. [PMID: 26556953 PMCID: PMC4628659 DOI: 10.1155/2015/272858] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/16/2015] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells form a semipermeable, regulated barrier that limits the passage of fluid, small molecules, and leukocytes between the bloodstream and the surrounding tissues. The adherens junction, a major mechanism of intercellular adhesion, is comprised of transmembrane cadherins forming homotypic interactions between adjacent cells and associated cytoplasmic catenins linking the cadherins to the cytoskeleton. Inflammatory conditions promote the disassembly of the adherens junction and a loss of intercellular adhesion, creating openings or gaps in the endothelium through which small molecules diffuse and leukocytes transmigrate. Tyrosine kinase signaling has emerged as a central regulator of the inflammatory response, partly through direct phosphorylation and dephosphorylation of the adherens junction components. This review discusses the findings that support and those that argue against a direct effect of cadherin and catenin phosphorylation in the disassembly of the adherens junction. Recent findings indicate a complex interaction between kinases, phosphatases, and the adherens junction components that allow a fine regulation of the endothelial permeability to small molecules, leukocyte migration, and barrier resealing.
Collapse
|
30
|
Chang YF, Hsu YF, Chiu PT, Huang WJ, Huang SW, Ou G, Sheu JR, Hsu MJ. WMJ-S-001, a novel aliphatic hydroxamate derivative, exhibits anti-angiogenic activities via Src-homology-2-domain-containing protein tyrosine phosphatase 1. Oncotarget 2015; 6:85-100. [PMID: 25415226 PMCID: PMC4381580 DOI: 10.18632/oncotarget.2765] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 11/15/2014] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis, one of the major routes for tumor invasion and metastasis represents a rational target for therapeutic intervention. Recent development in drug discovery has highlighted the diverse biological and pharmacological properties of hydroxamate derivatives. In this study, we characterized the anti-angiogenic activities of a novel aliphatic hydroxamate, WMJ-S-001, in an effort to develop novel angiogenesis inhibitors. WMJ-S-001 inhibited vascular endothelial growth factor (VEGF)-A-induced proliferation, invasion and endothelial tube formation of human umbilical endothelial cells (HUVECs). WMJ-S-001 suppressed VEGF-A-induced microvessel sprouting from aortic rings, and attenuated angiogenesis in in vivo mouse xenograft models. In addition, WMJ-S-001 inhibited the phosphorylations of VEGFR2, Src, FAK, Akt and ERK in VEGF-A-stimulated HUVECs. WMJ-S-001 caused an increase in SHP-1 phosphatase activity, whereas NSC-87877, a SHP-1 inhibitor, restored WMJ-S-001 suppression of VEGFR2 phosphorylation and cell proliferation. Furthermore, WMJ-S-001 inhibited cell cycle progression and induced cell apoptosis in HUVECs. These results are associated with p53 phosphorylation and acetylation and the modulation of p21 and survivin. Taken together, WMJ-S-001 was shown to modulate vascular endothelial cell remodeling through inhibiting VEGFR2 signaling and induction of apoptosis. These results also support the role of WMJ-S-001 as a potential drug candidate and warrant the clinical development in the treatment of cancer.
Collapse
Affiliation(s)
- Yi-Fang Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. Division of Hematology and Oncology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ya-Fen Hsu
- Division of General Surgery, Department of Surgery, Landseed Hospital, Taoyuan, Taiwan
| | - Pei-Ting Chiu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Jan Huang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Shiu-Wen Huang
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - George Ou
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joen-Rong Sheu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan. Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jen Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan. Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
31
|
Charming neighborhoods on the cell surface: plasma membrane microdomains regulate receptor tyrosine kinase signaling. Cell Signal 2015; 27:1963-76. [PMID: 26163824 DOI: 10.1016/j.cellsig.2015.07.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/07/2015] [Indexed: 12/14/2022]
Abstract
Receptor tyrosine kinases (RTK) are an important family of growth factor and hormone receptors that regulate many aspects of cellular physiology. Ligand binding by RTKs at the plasma membrane elicits activation of many signaling intermediates. The spatial and temporal regulation of RTK signaling within cells is an important determinant of receptor signaling outcome. In particular, the compartmentalization of the plasma membrane into a number of microdomains allows context-specific control of RTK signaling. Indeed various RTKs are recruited to and enriched within specific plasma membrane microdomains under various conditions, including lipid-ordered domains such as caveolae and lipid rafts, clathrin-coated structures, tetraspanin-enriched microdomains, and actin-dependent protrusive membrane microdomains such as dorsal ruffles and invadosomes. We examine the evidence for control of RTK signaling by each of these plasma membrane microdomains, as well as molecular mechanisms for how this spatial organization controls receptor signaling.
Collapse
|
32
|
The protein tyrosine phosphatase DEP-1/PTPRJ promotes breast cancer cell invasion and metastasis. Oncogene 2015; 34:5536-47. [DOI: 10.1038/onc.2015.9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 12/16/2014] [Accepted: 01/14/2015] [Indexed: 12/16/2022]
|
33
|
Takahashi K, Matafonov A, Sumarriva K, Ito H, Lauhan C, Zemel D, Tsuboi N, Chen J, Reynolds A, Takahashi T. CD148 tyrosine phosphatase promotes cadherin cell adhesion. PLoS One 2014; 9:e112753. [PMID: 25386896 PMCID: PMC4227875 DOI: 10.1371/journal.pone.0112753] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 10/14/2014] [Indexed: 01/06/2023] Open
Abstract
CD148 is a transmembrane tyrosine phosphatase that is expressed at cell junctions. Recent studies have shown that CD148 associates with the cadherin/catenin complex and p120 catenin (p120) may serve as a substrate. However, the role of CD148 in cadherin cell-cell adhesion remains unknown. Therefore, here we addressed this issue using a series of stable cells and cell-based assays. Wild-type (WT) and catalytically inactive (CS) CD148 were introduced to A431D (lacking classical cadherins), A431D/E-cadherin WT (expressing wild-type E-cadherin), and A431D/E-cadherin 764AAA (expressing p120-uncoupled E-cadherin mutant) cells. The effects of CD148 in cadherin adhesion were assessed by Ca2+ switch and cell aggregation assays. Phosphorylation of E-cadherin/catenin complex and Rho family GTPase activities were also examined. Although CD148 introduction did not alter the expression levels and complex formation of E-cadherin, p120, and β-catenin, CD148 WT, but not CS, promoted cadherin contacts and strengthened cell-cell adhesion in A431D/E-cadherin WT cells. This effect was accompanied by an increase in Rac1, but not RhoA and Cdc42, activity and largely diminished by Rac1 inhibition. Further, we demonstrate that CD148 reduces the tyrosine phosphorylation of p120 and β-catenin; causes the dephosphorylation of Y529 suppressive tyrosine residue in Src, a well-known CD148 site, increasing Src activity and enhancing the phosphorylation of Y228 (a Src kinase site) in p120, in E-cadherin contacts. Consistent with these findings, CD148 dephosphorylated both p120 and β-catenin in vitro. The shRNA-mediated CD148 knockdown in A431 cells showed opposite effects. CD148 showed no effects in A431D and A431D/E-cadherin 764AAA cells. In aggregate, these findings provide the first evidence that CD148 promotes E-cadherin adhesion by regulating Rac1 activity concomitant with modulation of p120, β-catenin, and Src tyrosine phosphorylation. This effect requires E-cadherin and p120 association.
Collapse
Affiliation(s)
- Keiko Takahashi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Anton Matafonov
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Katherine Sumarriva
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Hideyuki Ito
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Colette Lauhan
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Dana Zemel
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Nobuo Tsuboi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Jin Chen
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Albert Reynolds
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Takamune Takahashi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
- * E-mail:
| |
Collapse
|
34
|
Identification of receptor protein tyrosine phosphatases (RPTPs) as regulators of receptor tyrosine kinases (RTKs) using an RPTP siRNA-RTK substrate screen. Methods Mol Biol 2014; 1233:111-20. [PMID: 25319894 DOI: 10.1007/978-1-4939-1789-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Receptor tyrosine kinase (RTK) signaling exists in equilibrium between RTK tyrosyl phosphorylation and RTK tyrosyl dephosphorylation. Despite a detailed understanding of RTK tyrosyl phosphorylation, much less is known about RTK tyrosyl dephosphorylation. The receptor PTPs (RPTPs) are outstanding targets for the dephosphorylation of RTKs because of their mutual membrane proximity. In this chapter, we describe how to identify RPTPs that modulate the activity of RTKs using a siRNA screen and commercially available proteomic applications. The validation of putative RTKs as RPTP substrates using substrate-trapping approaches is detailed.
Collapse
|
35
|
Du Y, Grandis JR. Receptor-type protein tyrosine phosphatases in cancer. CHINESE JOURNAL OF CANCER 2014; 34:61-9. [PMID: 25322863 PMCID: PMC4360074 DOI: 10.5732/cjc.014.10146] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Protein tyrosine phosphatases (PTPs) play an important role in regulating cell signaling events in coordination with tyrosine kinases to control cell proliferation, apoptosis, survival, migration, and invasion. Receptor-type protein tyrosine phosphatases (PTPRs) are a subgroup of PTPs that share a transmembrane domain with resulting similarities in function and target specificity. In this review, we summarize genetic and epigenetic alterations including mutation, deletion, amplification, and promoter methylation of PTPRs in cancer and consider the consequences of PTPR alterations in different types of cancers. We also summarize recent developments using PTPRs as prognostic or predictive biomarkers and/or direct targets. Increased understanding of the role of PTPRs in cancer may provide opportunities to improve therapeutic approaches.
Collapse
Affiliation(s)
- Yu Du
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
36
|
Spring K, Lapointe L, Caron C, Langlois S, Royal I. Phosphorylation of DEP-1/PTPRJ on threonine 1318 regulates Src activation and endothelial cell permeability induced by vascular endothelial growth factor. Cell Signal 2014; 26:1283-93. [DOI: 10.1016/j.cellsig.2014.02.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 02/18/2014] [Indexed: 12/23/2022]
|
37
|
Frijhoff J, Dagnell M, Godfrey R, Ostman A. Regulation of protein tyrosine phosphatase oxidation in cell adhesion and migration. Antioxid Redox Signal 2014; 20:1994-2010. [PMID: 24111825 DOI: 10.1089/ars.2013.5643] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
SIGNIFICANCE Redox-regulated control of protein tyrosine phosphatases (PTPs) through inhibitory reversible oxidation of their active site is emerging as a novel and general mechanism for control of cell surface receptor-activated signaling. This mechanism allows for a previously unrecognized crosstalk between redox regulators and signaling pathways, governed by, for example, receptor tyrosine kinases and integrins, which control cell proliferation and migration. RECENT ADVANCES A large number of different molecules, in addition to hydrogen peroxide, have been found to induce PTP inactivation, including lipid peroxides, reactive nitrogen species, and hydrogen sulfide. Characterization of oxidized PTPs has identified different types of oxidative modifications that are likely to display differential sensitivity to various reducing systems. Accumulating evidence demonstrates that PTP oxidation occurs in a temporally and spatially restricted manner. Studies in cell and animal models indicate altered PTP oxidation in models of common diseases, such as cancer and metabolic/cardiovascular disease. Novel methods have appeared that allow characterization of global PTP oxidation. CRITICAL ISSUES As the understanding of the molecular and cellular biology of PTP oxidation is developing, it will be important to establish experimental procedures that allow analyses of PTP oxidation, and its regulation, in physiological and pathophysiological settings. Future studies should also aim to establish specific connections between various oxidants, specific PTPs, and defined signaling contexts. FUTURE DIRECTIONS Modulation of PTP activity still appears as a valid strategy for correction or inhibition of dys-regulated cell signaling. Continued studies on PTP oxidation might present yet unrecognized means to exploit this regulatory mechanism for pharmacological purposes.
Collapse
Affiliation(s)
- Jeroen Frijhoff
- 1 Department of Oncology-Pathology, Karolinska Institutet , Stockholm, Sweden
| | | | | | | |
Collapse
|
38
|
Brandes RP, Weissmann N, Schröder K. Redox-mediated signal transduction by cardiovascular Nox NADPH oxidases. J Mol Cell Cardiol 2014; 73:70-9. [PMID: 24560815 DOI: 10.1016/j.yjmcc.2014.02.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 11/30/2022]
Abstract
The only known function of the Nox family of NADPH oxidases is the production of reactive oxygen species (ROS). Some Nox enzymes show high tissue-specific expression and the ROS locally produced are required for synthesis of hormones or tissue components. In the cardiovascular system, Nox enzymes are low abundant and function as redox-modulators. By reacting with thiols, nitric oxide (NO) or trace metals, Nox-derived ROS elicit a plethora of cellular responses required for physiological growth factor signaling and the induction and adaptation to pathological processes. The interactions of Nox-derived ROS with signaling elements in the cardiovascular system are highly diverse and will be detailed in this article, which is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".
Collapse
Affiliation(s)
- Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Germany.
| | - Norbert Weissmann
- Giessen University Lung Center, Justus-Liebig-Universität, Gießen, Germany
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Germany
| |
Collapse
|
39
|
Anti-cancer activity of an osthole derivative, NBM-T-BMX-OS01: targeting vascular endothelial growth factor receptor signaling and angiogenesis. PLoS One 2013; 8:e81592. [PMID: 24312323 PMCID: PMC3842266 DOI: 10.1371/journal.pone.0081592] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 10/15/2013] [Indexed: 12/27/2022] Open
Abstract
Angiogenesis occurs during tissue growth, development and wound healing. It is also required for tumor progression and represents a rational target for therapeutic intervention. NBM-T-BMX-OS01 (BMX), derived from the semisynthesis of osthole, an active ingredient isolated from Chinese herb Cnidium monnieri (L.) Cuss., was recently shown to enhance learning and memory in rats. In this study, we characterized the anti-angiogenic activities of NBM-T-BMX-OS01 (BMX) in an effort to develop novel inhibitors to suppress angiogenesis and tumor growth. BMX inhibited vascular endothelial growth factor (VEGF)-induced proliferation, migration and endothelial tube formation in human umbilical endothelial cells (HUVECs). BMX also attenuated VEGF-induced microvessel sprouting from aortic rings ex vivo and reduced HCT116 colorectal cancer cells-induced angiogenesis in vivo. Moreover, BMX inhibited the phosphorylation of VEGFR2, FAK, Akt and ERK in HUVECs exposed to VEGF. BMX was also shown to inhibit HCT116 cell proliferation and to suppress the growth of subcutaneous xenografts of HCT116 cells in vivo. Taken together, this study provides evidence that BMX modulates vascular endothelial cell remodeling and leads to the inhibition of tumor angiogenesis. These results also support the role of BMX as a potential drug candidate and warrant the clinical development in the treatment of cancer.
Collapse
|
40
|
Nie L, Guo X, Esmailzadeh L, Zhang J, Asadi A, Collinge M, Li X, Kim JD, Woolls M, Jin SW, Dubrac A, Eichmann A, Simons M, Bender JR, Sadeghi MM. Transmembrane protein ESDN promotes endothelial VEGF signaling and regulates angiogenesis. J Clin Invest 2013; 123:5082-97. [PMID: 24177422 DOI: 10.1172/jci67752] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 08/29/2013] [Indexed: 12/21/2022] Open
Abstract
Aberrant blood vessel formation contributes to a wide variety of pathologies, and factors that regulate angiogenesis are attractive therapeutic targets. Endothelial and smooth muscle cell-derived neuropilin-like protein (ESDN) is a neuropilin-related transmembrane protein expressed in ECs; however, its potential effect on VEGF responses remains undefined. Here, we generated global and EC-specific Esdn knockout mice and demonstrated that ESDN promotes VEGF-induced human and murine EC proliferation and migration. Deletion of Esdn in the mouse interfered with adult and developmental angiogenesis, and knockdown of the Esdn homolog (dcbld2) in zebrafish impaired normal vascular development. Loss of ESDN in ECs blunted VEGF responses in vivo and attenuated VEGF-induced VEGFR-2 signaling without altering VEGF receptor or neuropilin expression. Finally, we found that ESDN associates with VEGFR-2 and regulates its complex formation with negative regulators of VEGF signaling, protein tyrosine phosphatases PTP1B and TC-PTP, and VE-cadherin. These findings establish ESDN as a regulator of VEGF responses in ECs that acts through a mechanism distinct from neuropilins. As such, ESDN may serve as a therapeutic target for angiogenesis regulation.
Collapse
MESH Headings
- Animals
- Antigens, CD/physiology
- Blood Vessels/embryology
- Cadherins/physiology
- Cells, Cultured
- Ear, External/blood supply
- Endothelium, Vascular/physiology
- Hindlimb/blood supply
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Ischemia/physiopathology
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Physiologic/physiology
- Neuropilins/physiology
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology
- RNA Interference
- RNA, Small Interfering/pharmacology
- Retinal Vessels/growth & development
- Vascular Endothelial Growth Factor A/physiology
- Vascular Endothelial Growth Factor Receptor-2/physiology
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish Proteins/physiology
Collapse
|
41
|
McCole DF. Phosphatase regulation of intercellular junctions. Tissue Barriers 2013; 1:e26713. [PMID: 24868494 DOI: 10.4161/tisb.26713] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 10/03/2013] [Accepted: 10/04/2013] [Indexed: 02/06/2023] Open
Abstract
Intercellular junctions represent the key contact points and sites of communication between neighboring cells. Assembly of these junctions is absolutely essential for the structural integrity of cell monolayers, tissues and organs. Disruption of junctions can have severe consequences such as diarrhea, edema and sepsis, and contribute to the development of chronic inflammatory diseases. Cell junctions are not static structures, but rather they represent highly dynamic micro-domains that respond to signals from the intracellular and extracellular environments to modify their composition and function. This review article will focus on the regulation of tight junctions and adherens junctions by phosphatase enzymes that play an essential role in preserving and modulating the properties of intercellular junction proteins.
Collapse
Affiliation(s)
- Declan F McCole
- Division of Biomedical Sciences; University of California, Riverside; Riverside, CA USA
| |
Collapse
|
42
|
Tan WH, Popel AS, Mac Gabhann F. Computational model of VEGFR2 pathway to ERK activation and modulation through receptor trafficking. Cell Signal 2013; 25:2496-510. [PMID: 23993967 DOI: 10.1016/j.cellsig.2013.08.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/24/2013] [Indexed: 01/09/2023]
Abstract
Vascular Endothelial Growth Factor (VEGF) signal transduction is central to angiogenesis in development and in pathological conditions such as cancer, retinopathy and ischemic diseases. We constructed and validated a computational model of VEGFR2 trafficking and signaling, to study the role of receptor trafficking kinetics in modulating ERK phosphorylation in VEGF-stimulated endothelial cells. Trafficking parameters were optimized and validated against four previously published in vitro experiments. Based on these parameters, model simulations demonstrated interesting behaviors that may be highly relevant to understanding VEGF signaling in endothelial cells. First, at moderate VEGF doses, VEGFR2 phosphorylation and ERK phosphorylation are related in a log-linear fashion, with a stable duration of ERK activation; but with higher VEGF stimulation, phosphoERK becomes saturated, and its duration increases. Second, a large endosomal fraction of VEGFR2 makes the ERK activation reaction network less sensitive to perturbations in VEGF dosage. Third, extracellular-matrix-bound VEGF binds and activates VEGFR2, but by internalizing at a slower rate, matrix-bound VEGF-induced intracellular ERK phosphorylation is predicted to be greater in magnitude and more sustained, in agreement with experimental evidence. Fourth, different endothelial cell types appear to have different trafficking rates, which result in different levels of endosomal receptor localization and different ERK response profiles.
Collapse
Affiliation(s)
- Wan Hua Tan
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Ave., Baltimore, MD 21205, USA
| | | | | |
Collapse
|
43
|
Knockout of Density-Enhanced Phosphatase-1 impairs cerebrovascular reserve capacity in an arteriogenesis model in mice. BIOMED RESEARCH INTERNATIONAL 2013; 2013:802149. [PMID: 24027763 PMCID: PMC3763586 DOI: 10.1155/2013/802149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/27/2013] [Accepted: 07/17/2013] [Indexed: 01/07/2023]
Abstract
Collateral growth, arteriogenesis, represents a proliferative mechanism involving endothelial cells, smooth muscle cells, and monocytes/macrophages. Here we investigated the role of Density-Enhanced Phosphatase-1 (DEP-1) in arteriogenesis in vivo, a protein-tyrosine-phosphatase that has controversially been discussed with regard to vascular cell biology. Wild-type C57BL/6 mice subjected to permanent left common carotid artery occlusion (CCAO) developed a significant diameter increase in distinct arteries of the circle of Willis, especially in the anterior cerebral artery. Analyzing the impact of loss of DEP-1 function, induction of collateralization was quantified after CCAO and hindlimb femoral artery ligation comparing wild-type and DEP-1−/− mice. Both cerebral collateralization assessed by latex perfusion and peripheral vessel growth in the femoral artery determined by microsphere perfusion and micro-CT analysis were not altered in DEP-1−/− compared to wild-type mice. Cerebrovascular reserve capacity, however, was significantly impaired in DEP-1−/− mice. Cerebrovascular transcriptional analysis of proarteriogenic growth factors and receptors showed specifically reduced transcripts of PDGF-B. SiRNA knockdown of DEP-1 in endothelial cells in vitro also resulted in significant PDGF-B downregulation, providing further evidence for DEP-1 in PDGF-B gene regulation. In summary, our data support the notion of DEP-1 as positive functional regulator in vascular cerebral arteriogenesis, involving differential PDGF-B gene expression.
Collapse
|
44
|
Lee HKP, Cording A, Vielmetter J, Zinn K. Interactions between a receptor tyrosine phosphatase and a cell surface ligand regulate axon guidance and glial-neuronal communication. Neuron 2013; 78:813-26. [PMID: 23764287 DOI: 10.1016/j.neuron.2013.04.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2013] [Indexed: 12/31/2022]
Abstract
We developed a screening method for orphan receptor ligands, in which cell-surface proteins are expressed in Drosophila embryos from GAL4-dependent insertion lines and ligand candidates identified by the presence of ectopic staining with receptor fusion proteins. Stranded at second (Sas) binds to the receptor tyrosine phosphatase Ptp10D in embryos and in vitro. Sas and Ptp10D can interact in trans when expressed in cultured cells. Interactions between Sas and Ptp10D on longitudinal axons are required to prevent them from abnormally crossing the midline. Sas is expressed on both neurons and glia, whereas Ptp10D is restricted to CNS axons. We conducted epistasis experiments by overexpressing Sas in glia and examining how the resulting phenotypes are changed by removal of Ptp10D from neurons. We find that neuronal Ptp10D restrains signaling by overexpressed glial Sas, which would otherwise produce strong glial and axonal phenotypes.
Collapse
Affiliation(s)
- Hyung-Kook Peter Lee
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | | |
Collapse
|
45
|
Tan WH, Popel AS, Mac Gabhann F. Computational Model of Gab1/2-Dependent VEGFR2 Pathway to Akt Activation. PLoS One 2013; 8:e67438. [PMID: 23805312 PMCID: PMC3689841 DOI: 10.1371/journal.pone.0067438] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 05/20/2013] [Indexed: 11/18/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) signal transduction is central to angiogenesis in development and in pathological conditions such as cancer, retinopathy and ischemic diseases. However, no detailed mass-action models of VEGF receptor signaling have been developed. We constructed and validated the first computational model of VEGFR2 trafficking and signaling, to study the opposing roles of Gab1 and Gab2 in regulation of Akt phosphorylation in VEGF-stimulated endothelial cells. Trafficking parameters were optimized against 5 previously published in vitro experiments, and the model was validated against six independent published datasets. The model showed agreement at several key nodes, involving scaffolding proteins Gab1, Gab2 and their complexes with Shp2. VEGFR2 recruitment of Gab1 is greater in magnitude, slower, and more sustained than that of Gab2. As Gab2 binds VEGFR2 complexes more transiently than Gab1, VEGFR2 complexes can recycle and continue to participate in other signaling pathways. Correspondingly, the simulation results show a log-linear relationship between a decrease in Akt phosphorylation and Gab1 knockdown while a linear relationship was observed between an increase in Akt phosphorylation and Gab2 knockdown. Global sensitivity analysis demonstrated the importance of initial-concentration ratios of antagonistic molecular species (Gab1/Gab2 and PI3K/Shp2) in determining Akt phosphorylation profiles. It also showed that kinetic parameters responsible for transient Gab2 binding affect the system at specific nodes. This model can be expanded to study multiple signaling contexts and receptor crosstalk and can form a basis for investigation of therapeutic approaches, such as tyrosine kinase inhibitors (TKIs), overexpression of key signaling proteins or knockdown experiments.
Collapse
Affiliation(s)
- Wan Hua Tan
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | | | | |
Collapse
|
46
|
Katsumoto TR, Kudo M, Chen C, Sundaram A, Callahan EC, Zhu JW, Lin J, Rosen CE, Manz BN, Lee JW, Matthay MA, Huang X, Sheppard D, Weiss A. The phosphatase CD148 promotes airway hyperresponsiveness through SRC family kinases. J Clin Invest 2013; 123:2037-48. [PMID: 23543053 DOI: 10.1172/jci66397] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 02/07/2013] [Indexed: 01/10/2023] Open
Abstract
Increased airway smooth muscle (ASM) contractility and the development of airway hyperresponsiveness (AHR) are cardinal features of asthma, but the signaling pathways that promote these changes are poorly understood. Tyrosine phosphorylation is tightly regulated by the opposing actions of protein tyrosine kinases and phosphatases, but little is known about whether tyrosine phosphatases influence AHR. Here, we demonstrate that genetic inactivation of receptor-like protein tyrosine phosphatase J (Ptprj), which encodes CD148, protected mice from the development of increased AHR in two different asthma models. Surprisingly, CD148 deficiency minimally affected the inflammatory response to allergen, but significantly altered baseline pulmonary resistance. Mice specifically lacking CD148 in smooth muscle had decreased AHR, and the frequency of calcium oscillations in CD148-deficient ASM was substantially attenuated, suggesting that signaling pathway alterations may underlie ASM contractility. Biochemical analysis of CD148-deficient ASM revealed hyperphosphorylation of the C-terminal inhibitory tyrosine of SRC family kinases (SFKs), implicating CD148 as a critical positive regulator of SFK signaling in ASM. The effect of CD148 deficiency on ASM contractility could be mimicked by treatment of both mouse trachea and human bronchi with specific SFK inhibitors. Our studies identify CD148 and the SFKs it regulates in ASM as potential targets for the treatment of AHR.
Collapse
Affiliation(s)
- Tamiko R Katsumoto
- Division of Rheumatology and Rosalind Russell Medical Research Center for Arthritis, University of California San Francisco (UCSF), San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee H, Bennett AM. Receptor protein tyrosine phosphatase-receptor tyrosine kinase substrate screen identifies EphA2 as a target for LAR in cell migration. Mol Cell Biol 2013; 33:1430-41. [PMID: 23358419 PMCID: PMC3624262 DOI: 10.1128/mcb.01708-12] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 01/23/2013] [Indexed: 01/08/2023] Open
Abstract
Receptor tyrosine kinases (RTKs) exist in equilibrium between tyrosyl-phosphorylated and dephosphorylated states. Despite a detailed understanding of how RTKs become tyrosyl phosphorylated, much less is known about RTK tyrosyl dephosphorylation. Receptor protein tyrosine phosphatases (RPTPs) can play essential roles in the dephosphorylation of RTKs. However, a complete understanding of the involvement of the RPTP subfamily in RTK tyrosyl dephosphorylation has not been established. In this study, we have employed a small interfering RNA (siRNA) screen to identify RPTPs in the human genome that serve as RTK phosphatases. We observed that each RPTP induced a unique fingerprint of tyrosyl phosphorylation among 42 RTKs. We identified EphA2 as a novel LAR substrate. LAR dephosphorylated EphA2 at phosphotyrosyl 930, uncoupling Nck1 from EphA2 and thereby attenuating EphA2-mediated cell migration. These results demonstrate that each RPTP exerts a unique regulatory fingerprint of RTK tyrosyl dephosphorylation and suggest a complex signaling interplay between RTKs and RPTPs. Furthermore, we observed that LAR modulates cell migration through EphA2 site-specific dephosphorylation.
Collapse
Affiliation(s)
| | - Anton M. Bennett
- Department of Pharmacology
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
48
|
|
49
|
Paduano F, Ortuso F, Campiglia P, Raso C, Iaccino E, Gaspari M, Gaudio E, Mangone G, Carotenuto A, Bilotta A, Narciso D, Palmieri C, Agosti V, Artese A, Gomez-Monterrey I, Sala M, Cuda G, Iuliano R, Perrotti N, Scala G, Viglietto G, Alcaro S, Croce CM, Novellino E, Fusco A, Trapasso F. Isolation and functional characterization of peptide agonists of PTPRJ, a tyrosine phosphatase receptor endowed with tumor suppressor activity. ACS Chem Biol 2012; 7:1666-76. [PMID: 22759068 DOI: 10.1021/cb300281t] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PTPRJ is a receptor-type protein tyrosine phosphatase whose expression is strongly reduced in the majority of investigated cancer cell lines and tumor specimens. PTPRJ negatively interferes with mitogenic signals originating from several oncogenic receptor tyrosine kinases, including HGFR, PDGFR, RET, and VEGFR-2. Here we report the isolation and characterization of peptides from a random peptide phage display library that bind and activate PTPRJ. These agonist peptides, which are able to both circularize and form dimers in acqueous solution, were assayed for their biochemical and biological activity on both human cancer cells and primary endothelial cells (HeLa and HUVEC, respectively). Our results demonstrate that binding of PTPRJ-interacting peptides to cell cultures dramatically reduces the extent of both MAPK phosphorylation and total phosphotyrosine levels; conversely, they induce a significant increase of the cell cycle inhibitor p27(Kip1). Moreover, PTPRJ agonist peptides both reduce proliferation and trigger apoptosis of treated cells. Our data indicate that peptide agonists of PTPRJ positively modulate the PTPRJ activity and may lead to novel targeted anticancer therapies.
Collapse
Affiliation(s)
- Francesco Paduano
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Francesco Ortuso
- Laboratorio
di Chimica Farmaceutica
Computazionale, Dipartimento di Scienze Farmacobiologiche, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100 Catanzaro, Italy
| | - Pietro Campiglia
- Dipartimento di Scienze Farmaceutiche
e Biomediche, Sezione Chimico-Tecnologica, Università di Salerno, 84084 Fisciano (Salerno), Italy
| | - Cinzia Raso
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Enrico Iaccino
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Marco Gaspari
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Eugenio Gaudio
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
- Department of Molecular Virology,
Immunology and Medical Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United
States
| | - Graziella Mangone
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Alfonso Carotenuto
- Dipartimento di
Chimica Farmaceutica
e Tossicologica, Università degli Studi di Napoli “Federico II”, 80131 Naples, Italy
| | - Anna Bilotta
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Domenico Narciso
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Camillo Palmieri
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Valter Agosti
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Anna Artese
- Laboratorio
di Chimica Farmaceutica
Computazionale, Dipartimento di Scienze Farmacobiologiche, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100 Catanzaro, Italy
| | - Isabel Gomez-Monterrey
- Dipartimento di
Chimica Farmaceutica
e Tossicologica, Università degli Studi di Napoli “Federico II”, 80131 Naples, Italy
| | - Marina Sala
- Dipartimento di Scienze Farmaceutiche
e Biomediche, Sezione Chimico-Tecnologica, Università di Salerno, 84084 Fisciano (Salerno), Italy
| | - Giovanni Cuda
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Rodolfo Iuliano
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Nicola Perrotti
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Giuseppe Scala
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Giuseppe Viglietto
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| | - Stefano Alcaro
- Laboratorio
di Chimica Farmaceutica
Computazionale, Dipartimento di Scienze Farmacobiologiche, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100 Catanzaro, Italy
| | - Carlo M. Croce
- Department of Molecular Virology,
Immunology and Medical Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United
States
| | - Ettore Novellino
- Dipartimento di
Chimica Farmaceutica
e Tossicologica, Università degli Studi di Napoli “Federico II”, 80131 Naples, Italy
| | - Alfredo Fusco
- Dipartimento di Biologia e Patologia
Cellulare e Molecolare c/o Istituto di Endocrinologia ed Oncologia
Sperimentale del CNR, Università degli Studi di Napoli “Federico II”, 80131 Naples, Italy
| | - Francesco Trapasso
- Dipartimento di Medicina Sperimentale
e Clinica, Università “Magna Græcia” di Catanzaro, Campus “S. Venuta”, 88100
Catanzaro, Italy
| |
Collapse
|
50
|
Saul VV, de la Vega L, Milanovic M, Krüger M, Braun T, Fritz-Wolf K, Becker K, Schmitz ML. HIPK2 kinase activity depends on cis-autophosphorylation of its activation loop. J Mol Cell Biol 2012; 5:27-38. [PMID: 23000554 DOI: 10.1093/jmcb/mjs053] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The multitude of mechanisms regulating the activity of protein kinases includes phosphorylation of amino acids contained in the activation loop. Here we show that the serine/threonine kinase HIPK2 (homeodomain-interacting protein kinase 2) is heavily modified by autophosphorylation, which occurs by cis-autophosphorylation at the activation loop and by trans-autophosphorylation at other phosphorylation sites. Cis-autophosphorylation of HIPK2 at Y354 and S357 in the activation loop is essential for its kinase function and the binding to substrates and the interaction partner Pin1. HIPK2 activation loop phosphorylation is also required for its biological activity as a regulator of gene expression and cell proliferation. Phosphorylation of HIPK2 at Y354 alone is not sufficient for full HIPK2 activity, which is in marked contrast to some dual-specificity tyrosine-phosphorylated and regulated kinases where tyrosine phosphorylation is absolutely essential. This study shows that differential phosphorylation of HIPK2 provides a mechanism for controlling and specifying the signal output from this kinase.
Collapse
Affiliation(s)
- Vera V Saul
- Department of Biochemistry, Medical Faculty, Justus Liebig University, Member of the German Center for Lung Research, Friedrichstrasse 24, Giessen 35392, Germany
| | | | | | | | | | | | | | | |
Collapse
|