1
|
Du R, Wang P, Tian N. CD3ζ-Mediated Signaling Protects Retinal Ganglion Cells in Glutamate Excitotoxicity of the Retina. Cells 2024; 13:1006. [PMID: 38920637 PMCID: PMC11201742 DOI: 10.3390/cells13121006] [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: 03/30/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Excessive levels of glutamate activity could potentially damage and kill neurons. Glutamate excitotoxicity is thought to play a critical role in many CNS and retinal diseases. Accordingly, glutamate excitotoxicity has been used as a model to study neuronal diseases. Immune proteins, such as major histocompatibility complex (MHC) class I molecules and their receptors, play important roles in many neuronal diseases, while T-cell receptors (TCR) are the primary receptors of MHCI. We previously showed that a critical component of TCR, CD3ζ, is expressed by mouse retinal ganglion cells (RGCs). The mutation of CD3ζ or MHCI molecules compromises the development of RGC structure and function. In this study, we investigated whether CD3ζ-mediated molecular signaling regulates RGC death in glutamate excitotoxicity. We show that mutation of CD3ζ significantly increased RGC survival in NMDA-induced excitotoxicity. In addition, we found that several downstream molecules of TCR, including Src (proto-oncogene tyrosine-protein kinase) family kinases (SFKs) and spleen tyrosine kinase (Syk), are expressed by RGCs. Selective inhibition of an SFK member, Hck, or Syk members, Syk or Zap70, significantly increased RGC survival in NMDA-induced excitotoxicity. These results provide direct evidence to reveal the underlying molecular mechanisms that control RGC death under disease conditions.
Collapse
Affiliation(s)
- Rui Du
- Department of Ophthalmology and Visual Science, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (R.D.); (P.W.)
| | - Ping Wang
- Department of Ophthalmology and Visual Science, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (R.D.); (P.W.)
| | - Ning Tian
- Department of Ophthalmology and Visual Science, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (R.D.); (P.W.)
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84132, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84132, USA
- Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA
| |
Collapse
|
2
|
Britto-Júnior J, Furlaneto R, Lima AT, de Oliveira MG, Severino B, Frecentese F, Fiorino F, Caliendo G, Muscará MN, De Nucci G. GKT137831 and hydrogen peroxide increase the release of 6-nitrodopamine from the human umbilical artery, rat-isolated right atrium, and rat-isolated vas deferens. Front Pharmacol 2024; 15:1348876. [PMID: 38645555 PMCID: PMC11026650 DOI: 10.3389/fphar.2024.1348876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/01/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction: The human umbilical artery (HUA), rat-isolated right atrium, and rat-isolated vas deferens present a basal release of 6-nitrodopamine (6-ND). The basal release of 6-ND from these tissues was significantly decreased (but not abolished) when the tissues were pre-incubated with Nω-nitro-L-arginine methyl ester (L-NAME). Methods: In this study, the effect of the pharmacological modulation of the redox environment on the basal release of 6-ND was investigated. The basal release of 6-ND was measured using Liquid chromatography with tandem mass spectrometry (LC-MS/MS). Results and Discussion: Pre-incubation (30 min) of the tissues with GKT137831 (1 μM) caused a significant increase in the basal release of 6-ND from all tissues. In the HUA, pre-incubation with diphenyleneiodonium (DPI) (100 μM) also caused significant increases in the basal release of 6-ND. Preincubation of the HUA with hydrogen peroxide (H2O2) (100 μM) increased 6-ND basal release, whereas pre-incubation with catalase (1,000 U/mL) significantly decreased it. Pre-incubation of the HUA with superoxide dismutase (SOD) (250 U/mL; 30 min) also significantly increased the basal release of 6-ND. Preincubation of the HUA with either allopurinol (100 μM) or uric acid (1 mM) had no effect on the basal release of 6-ND. Pre-treatment of the HUA with L-NAME (100 μM) prevented the increase in the basal release of 6-ND induced by GKT137831, diphenyleneiodonium, and H2O2. The results obtained indicate a major role of endogenous H2O2 and peroxidases as modulators of 6- ND biosynthesis/release and a lack of peroxynitrite contribution.
Collapse
Affiliation(s)
- José Britto-Júnior
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Rafael Furlaneto
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Department of Pharmacology, Faculty of Medicine, Sao Leopoldo Mandic, Campinas, São Paulo, Brazil
| | - Antonio Tiago Lima
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Beatrice Severino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Francesco Frecentese
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Ferdinando Fiorino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Giuseppe Caliendo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Marcelo Nicolás Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), Sāo Paulo, Brazil
| | - Gilberto De Nucci
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), Sāo Paulo, Brazil
- Department of Pharmacology, Faculty of Medicine, Sao Leopoldo Mandic, Campinas, São Paulo, Brazil
| |
Collapse
|
3
|
Liu K, Hao Z, Zheng H, Wang H, Zhang L, Yan M, Tuerhong R, Zhou Y, Wang Y, Pang T, Shi L. Repurposing of rilpivirine for preventing platelet β3 integrin-dependent thrombosis by targeting c-Src active autophosphorylation. Thromb Res 2023; 229:53-68. [PMID: 37413892 DOI: 10.1016/j.thromres.2023.06.031] [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: 04/17/2023] [Revised: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND HIV-infected individuals are known to be at higher risk for thrombotic cardiovascular disease (CVD), which may also be differentially affected by components of anti-HIV drugs. To identify the effects of a series of FDA-approved anti-HIV drugs on platelet aggregation in humans, focusing on the novel pharmacological effects of rilpivirine (RPV), a reverse transcriptase inhibitor, on platelet function both in vitro and in vivo and the mechanisms involved. METHODS AND RESULTS In vitro studies showed that RPV was the only anti-HIV reagent that consistently and efficiently inhibited aggregation elicited by different agonists, exocytosis, morphological extension on fibrinogen, and clot retraction. Treatment of mice with RPV significantly prevented thrombus formation in FeCl3-injured mesenteric vessels, postcava with stenosis surgery, and ADP -induced pulmonary embolism models without defects in platelet viability, tail bleeding, and coagulation activities. RPV also improved cardiac performance in mice with post-ischemic reperfusion. A mechanistic study revealed that RPV preferentially attenuated fibrinogen-stimulated Tyr773 phosphorylation of β3-integrin by inhibiting Tyr419 autophosphorylation of c-Src. Molecular docking and surface plasmon resonance analyses showed that RPV can bind directly to c-Src. Further mutational analysis showed that the Phe427 residue of c-Src is critical for RPV interaction, suggesting a novel interaction site for targeting c-Src to block β3-integrin outside-in signaling. CONCLUSION These results demonstrated that RPV was able to prevent the progression of thrombotic CVDs by interrupting β3-integrin-mediated outside-in signaling via inhibiting c-Src activation without hemorrhagic side effects, highlighting RPV as a promising reagent for the prevention and therapy of thrombotic CVDs.
Collapse
Affiliation(s)
- Kui Liu
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Xiamen 361000, China; State Key Laboratory of Natural Medicines, New Drug Screening Center, Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China
| | - Zhen Hao
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Xiamen 361000, China; College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China
| | - Hao Zheng
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China
| | - Haojie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Luying Zhang
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China
| | - Minghui Yan
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China
| | - Reyisha Tuerhong
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China
| | - Yuling Zhou
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Xiamen 361000, China
| | - Yan Wang
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Xiamen 361000, China.
| | - Tao Pang
- State Key Laboratory of Natural Medicines, New Drug Screening Center, Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing 210009, China.
| | - Lei Shi
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Xiamen 361000, China; College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section, South Lv shun Road, Dalian 116044, China.
| |
Collapse
|
4
|
Development of 2'-aminospiro [pyrano[3,2-c]quinoline]-3'-carbonitrile derivatives as non-ATP competitive Src kinase inhibitors that suppress breast cancer cell migration and proliferation. Bioorg Chem 2021; 116:105344. [PMID: 34598088 DOI: 10.1016/j.bioorg.2021.105344] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/31/2021] [Accepted: 09/07/2021] [Indexed: 02/03/2023]
Abstract
Src kinase activity controls diverse cellular functions, including cell growth, migration, adhesion, and survival. It is de-regulated in several cancers, including breast cancer, where it is highly expressed and phosphorylated. Thus, targeting Src by a small molecule is a feasible strategy for managing different breast cancer types. Several Src kinase inhibitors are available, including the FDA-approved drug (dasatinib). However, they are primarily ATP-competitive inhibitors that have been reported to lack specificity towards Src. We have a long-time interest in discovering protein kinase inhibitors that are non-competitive for ATP. In this project, three groups of 2'-aminospiro[pyrano[3,2-c]quinoline]-3'-carbonitrile derivatives were designed and synthesized, hypothesizing that small molecules with a spiro scaffold appended to a pyrano[3,2-c]quinoline analog could act as non-ATP competitive Src kinase inhibitors. 3b, 3c, and 3d inhibited Src kinase activity with IC50s of 4.9, 5.9, and 0.9 μM, respectively. At the same time, they did not impact the MDM2/p53 interaction in HEK293 cells, which has been reported to be affected by some spirocyclic compounds. 25 µM of 3b, 3c, or 3d did not inhibit the kinase activity of ERK2, JNK1, or p38-alpha in an in-vitro kinase assay. Steady-state kinetic studies for the effect of 3d on the ability of recombinant Src to phosphorylate its substrate (Srctide) revealed a non-ATP competitive inhibition mechanism. 1.6 µM of 3d was enough to diminish Src, Fak, and paxillin phosphorylation in the breast cancer cell lines MDA-MB-231 and MCF7. In the NCI screening, 3d induced broad tumor cytotoxicity for the NCI-60 cell lines, including all the breast cancer cell lines. The potency of 3b, 3c, and 3d to inhibit migration, proliferation, and colony formation of MDA-MB-231 and proliferation of MCF7 cells correlates with their potency to suppress Src kinase activity in the same cell line. Noticeably, the cell growth suppression and apoptosis induction in the tested cell lines can be attributed to the ability of the new derivatives to suppress the ERK and Akt survival pathways downstream of Src.
Collapse
|
5
|
You KS, Yi YW, Cho J, Park JS, Seong YS. Potentiating Therapeutic Effects of Epidermal Growth Factor Receptor Inhibition in Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:589. [PMID: 34207383 PMCID: PMC8233743 DOI: 10.3390/ph14060589] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with aggressive characteristics and few therapeutic options. The lack of an appropriate therapeutic target is a challenging issue in treating TNBC. Although a high level expression of epidermal growth factor receptor (EGFR) has been associated with a poor prognosis among patients with TNBC, targeted anti-EGFR therapies have demonstrated limited efficacy for TNBC treatment in both clinical and preclinical settings. However, with the advantage of a number of clinically approved EGFR inhibitors (EGFRis), combination strategies have been explored as a promising approach to overcome the intrinsic resistance of TNBC to EGFRis. In this review, we analyzed the literature on the combination of EGFRis with other molecularly targeted therapeutics or conventional chemotherapeutics to understand the current knowledge and to provide potential therapeutic options for TNBC treatment.
Collapse
Affiliation(s)
- Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
| | - Yong Weon Yi
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeonghee Cho
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| |
Collapse
|
6
|
Li Y, Bao Y, Zheng H, Qin Y, Hua B. The nonreceptor protein tyrosine kinase Src participates in every step of cancer-induced bone pain. Biomed Pharmacother 2021; 141:111822. [PMID: 34147901 DOI: 10.1016/j.biopha.2021.111822] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/30/2021] [Accepted: 06/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer-induced bone pain (CIBP) is a refractory form of pain that has a high incidence in advanced tumors. Src protein tyrosine kinase is mainly composed of six domains, with two states of automatic inhibition and activation. The modular domain allows Src to conveniently regulate by and communicate with a variety of proteins, directly or indirectly participate in each step of the CIBP process. Src is beneficial to the growth and proliferation of tumor cells, and it can promote the metastases of primary tumors to bone. In the microenvironment of bone metastasis, it mainly mediates bone resorption, activates related peripheral receptors to participate in the formation of pain signals, and may promote the generation of pathological sensory nerve fibers. In the process of pain signal transmission, it mainly mediates NMDAR and central glial cells to regulate pain signal intensity and central sensitization, but it is not limited to these two aspects. Both basic experimentation and clinical research have shown encouraging potential, providing new ideas and inspiration for the prevention and treatment of CIBP.
Collapse
Affiliation(s)
- Yaoyuan Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanju Bao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yinggang Qin
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
7
|
Grayson JD, Baumgartner MP, Santos Souza CD, Dawes SJ, El Idrissi IG, Louth JC, Stimpson S, Mead E, Dunbar C, Wolak J, Sharman G, Evans D, Zhuravleva A, Roldan MS, Colabufo NA, Ning K, Garwood C, Thomas JA, Partridge BM, de la Vega de Leon A, Gillet VJ, Rauter AP, Chen B. Amyloid binding and beyond: a new approach for Alzheimer's disease drug discovery targeting Aβo-PrP C binding and downstream pathways. Chem Sci 2021; 12:3768-3785. [PMID: 34163650 PMCID: PMC8179515 DOI: 10.1039/d0sc04769d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/08/2021] [Indexed: 01/18/2023] Open
Abstract
Amyloid β oligomers (Aβo) are the main toxic species in Alzheimer's disease, which have been targeted for single drug treatment with very little success. In this work we report a new approach for identifying functional Aβo binding compounds. A tailored library of 971 fluorine containing compounds was selected by a computational method, developed to generate molecular diversity. These compounds were screened for Aβo binding by a combined 19F and STD NMR technique. Six hits were evaluated in three parallel biochemical and functional assays. Two compounds disrupted Aβo binding to its receptor PrPC in HEK293 cells. They reduced the pFyn levels triggered by Aβo treatment in neuroprogenitor cells derived from human induced pluripotent stem cells (hiPSC). Inhibitory effects on pTau production in cortical neurons derived from hiPSC were also observed. These drug-like compounds connect three of the pillars in Alzheimer's disease pathology, i.e. prion, Aβ and Tau, affecting three different pathways through specific binding to Aβo and are, indeed, promising candidates for further development.
Collapse
Affiliation(s)
- James D Grayson
- Department of Chemistry, University of Sheffield Brookhill Sheffield S3 7HF UK
| | - Matthew P Baumgartner
- Computational Chemistry and Cheminformatics, Eli Lilly and Company, Lilly Biotechnology Center San Diego CA 92121 USA
| | | | - Samuel J Dawes
- Department of Chemistry, University of Sheffield Brookhill Sheffield S3 7HF UK
- Faculty of Biological Sciences, University of Leeds Leeds LS2 9JT UK
| | | | - Jennifer C Louth
- Department of Chemistry, University of Sheffield Brookhill Sheffield S3 7HF UK
| | - Sasha Stimpson
- Department of Chemistry, University of Sheffield Brookhill Sheffield S3 7HF UK
| | - Emma Mead
- Computational Chemistry and Chemoinformatics, Eli Lilly and Company Erl Wood Windlesham GU20 6PH UK
| | - Charlotte Dunbar
- Computational Chemistry and Chemoinformatics, Eli Lilly and Company Erl Wood Windlesham GU20 6PH UK
| | - Joanna Wolak
- Computational Chemistry and Chemoinformatics, Eli Lilly and Company Erl Wood Windlesham GU20 6PH UK
| | - Gary Sharman
- Computational Chemistry and Chemoinformatics, Eli Lilly and Company Erl Wood Windlesham GU20 6PH UK
| | - David Evans
- Computational Chemistry and Chemoinformatics, Eli Lilly and Company Erl Wood Windlesham GU20 6PH UK
| | | | | | - Nicola Antonio Colabufo
- Univ Bari, Biofordrug Via Edoardo Orabona 4 I-70125 Bari Italy
- Univ Bari, Dipartimento Farm Sci Farmaco Via Edoardo Orabona 4 I-70125 Bari Italy
| | - Ke Ning
- Sheffield Institute of Translational Neuroscience, University of Sheffield Sheffield S10 2HQ UK
| | - Claire Garwood
- Sheffield Institute of Translational Neuroscience, University of Sheffield Sheffield S10 2HQ UK
| | - James A Thomas
- Department of Chemistry, University of Sheffield Brookhill Sheffield S3 7HF UK
| | | | | | | | - Amélia P Rauter
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa ED C8, 5 piso 1749-016 Lisboa Portugal
| | - Beining Chen
- Department of Chemistry, University of Sheffield Brookhill Sheffield S3 7HF UK
| |
Collapse
|
8
|
Wang Y, Shi Z, Zhang Y, Yan J, Yu W, Chen L. Oligomer β-amyloid Induces Hyperactivation of Ras to Impede NMDA Receptor-Dependent Long-Term Potentiation in Hippocampal CA1 of Mice. Front Pharmacol 2020; 11:595360. [PMID: 33536910 PMCID: PMC7848859 DOI: 10.3389/fphar.2020.595360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/20/2020] [Indexed: 12/02/2022] Open
Abstract
The activity of Ras, a small GTPase protein, is increased in brains with Alzheimer’s disease. The objective of this study was to determine the influence of oligomeric Aβ1-42 on the activation of Ras, and the involvement of the Ras hyperactivity in Aβ1-42-induced deficits in spatial cognition and hippocampal synaptic plasticity. Herein, we show that intracerebroventricular injection of Aβ1-42 in mice (Aβ-mice) enhanced hippocampal Ras activation and expression, while 60 min incubation of hippocampal slices in Aβ1-42 (Aβ-slices) only elevated Ras activity. Aβ-mice showed deficits in spatial cognition and NMDA receptor (NMDAR)-dependent long-term potentiation (LTP) in hippocampal CA1, but basal synaptic transmission was enhanced. The above effects of Aβ1-42 were corrected by the Ras inhibitor farnesylthiosalicylic acid (FTS). ERK2 phosphorylation increased, and Src phosphorylation decreased in Aβ-mice and Aβ1-42-slices. Both were corrected by FTS. In CA1 pyramidal cells of Aβ1-42-slices, the response of AMPA receptor and phosphorylation of GluR1 were enhanced with dependence on Ras activation rather than ERK signaling. In contrast, NMDA receptor (NMDAR) function and GluN2A/2B phosphorylation were downregulated in Aβ1-42-slices, which was recovered by application of FTS or the Src activator ouabain, and mimicked in control slices treated with the Src inhibitor PP2. The administration of PP2 impaired the spatial cognition and LTP induction in control mice and FTS-treated Aβ-mice. The treatment of Aβ-mice with ouabain rescued Aβ-impaired spatial cognition and LTP. Overall, the results indicate that the oligomeric Aβ1-42 hyperactivates Ras and thereby causes the downregulation of Src which impedes NMDAR-dependent LTP induction resulting in cognitive deficits.
Collapse
Affiliation(s)
- Ya Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Zhaochun Shi
- Department of Neurology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yajie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Jun Yan
- Department of Geriatric Medicine, Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenfeng Yu
- Key Laboratory of Endemic and Ethnic Diseases of Education Ministry, Guizhou Medical University, Guizhou, China
| | - Ling Chen
- Department of Physiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Mo GCH, Posner C, Rodriguez EA, Sun T, Zhang J. A rationally enhanced red fluorescent protein expands the utility of FRET biosensors. Nat Commun 2020; 11:1848. [PMID: 32296061 PMCID: PMC7160135 DOI: 10.1038/s41467-020-15687-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 03/20/2020] [Indexed: 11/30/2022] Open
Abstract
Genetically encoded Förster Resonance Energy Transfer (FRET)-based biosensors are powerful tools to illuminate spatiotemporal regulation of cell signaling in living cells, but the utility of the red spectrum for biosensing was limited due to a lack of bright and stable red fluorescent proteins. Here, we rationally improve the photophysical characteristics of the coral-derived fluorescent protein TagRFP-T. We show that a new single-residue mutant, super-TagRFP (stagRFP) has nearly twice the molecular brightness of TagRFP-T and negligible photoactivation. stagRFP facilitates significant improvements on multiple green-red biosensors as a FRET acceptor and is an efficient FRET donor that supports red/far-red FRET biosensing. Capitalizing on the ability of stagRFP to couple with multiple FRET partners, we develop a novel multiplex method to examine the confluence of signaling activities from three kinases simultaneously in single living cells, providing evidence for a role of Src family kinases in regulating growth factor induced Akt and ERK activities.
Collapse
Affiliation(s)
- Gary C H Mo
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Clara Posner
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Erik A Rodriguez
- Department of Chemistry, The George Washington University, Washington, DC, 20052, USA
| | - Tengqian Sun
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jin Zhang
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
10
|
Tsuyuguchi M, Nakaniwa T, Hirasawa A, Nakanishi I, Kinoshita T. Structural insights for producing CK2α1-specific inhibitors. Bioorg Med Chem Lett 2019; 30:126837. [PMID: 31859160 DOI: 10.1016/j.bmcl.2019.126837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/06/2019] [Accepted: 11/15/2019] [Indexed: 11/25/2022]
Abstract
Casein kinase 2 catalytic subunit (CK2α) is classified into two subtypes CK2α1 and CK2α2. CK2α1 is a drug discovery target, whereas CK2α2 is an off-target of CK2α1 inhibitors. High amino acid sequence homology between these subtypes hampers efforts to produce ATP competitive inhibitors that are highly selective to CK2α1. Hematein was identified previously as a non-ATP-competitive inhibitor for CK2α1, whereas this compound acts as an ATP competitive CK2α2 inhibitor. Crystal structures of CK2α1 and CK2α2 in complex with hematein revealed distinct binding features that provide structural insights for producing CK2α1-selective inhibitors.
Collapse
Affiliation(s)
- Masato Tsuyuguchi
- Graduate School of Science, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan.
| | - Tetsuko Nakaniwa
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Akira Hirasawa
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Isao Nakanishi
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, Osaka 577-8502, Japan
| | - Takayoshi Kinoshita
- Graduate School of Science, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| |
Collapse
|
11
|
Sun XD, Wang A, Ma P, Gong S, Tao J, Yu XM, Jiang X. Regulation of the firing activity by PKA-PKC-Src family kinases in cultured neurons of hypothalamic arcuate nucleus. J Neurosci Res 2019; 98:384-403. [PMID: 31407399 PMCID: PMC6916362 DOI: 10.1002/jnr.24516] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 07/18/2019] [Accepted: 07/30/2019] [Indexed: 12/19/2022]
Abstract
The cAMP‐dependent protein kinase A family (PKAs), protein kinase C family (PKCs), and Src family kinases (SFKs) are found to play important roles in pain hypersensitivity. However, more detailed investigations are still needed in order to understand the mechanisms underlying the actions of PKAs, PKCs, and SFKs. Neurons in the hypothalamic arcuate nucleus (ARC) are found to be involved in the regulation of pain hypersensitivity. Here we report that the action potential (AP) firing activity of ARC neurons in culture was up‐regulated by application of the adenylate cyclase activator forskolin or the PKC activator PMA, and that the forskolin or PMA application‐induced up‐regulation of AP firing activity could be blocked by pre‐application of the SFK inhibitor PP2. SFK activation also up‐regulated the AP firing activity and this effect could be prevented by pre‐application of the inhibitors of PKCs, but not of PKAs. Furthermore, we identified that forskolin or PMA application caused increases in the phosphorylation not only in PKAs at T197 or PKCs at S660 and PKCα/βII at T638/641, but also in SFKs at Y416. The forskolin or PMA application‐induced increase in the phosphorylation of PKAs or PKCs was not affected by pre‐treatment with PP2. The regulations of the SFK and AP firing activities by PKCs were independent upon the translocation of either PKCα or PKCβII. Thus, it is demonstrated that PKAs may act as an upstream factor(s) to enhance SFKs while PKCs and SFKs interact reciprocally, and thereby up‐regulate the AP firing activity in hypothalamic ARC neurons.
Collapse
Affiliation(s)
- Xiao-Dong Sun
- Key Laboratory of Pain Basic Research and Clinical Therapy, Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, China
| | - Anqi Wang
- Key Laboratory of Pain Basic Research and Clinical Therapy, Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, China
| | - Peng Ma
- Key Laboratory of Pain Basic Research and Clinical Therapy, Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, China
| | - Shan Gong
- Key Laboratory of Pain Basic Research and Clinical Therapy, Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, China
| | - Jin Tao
- Key Laboratory of Pain Basic Research and Clinical Therapy, Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, China
| | - Xian-Min Yu
- Key Laboratory of Pain Basic Research and Clinical Therapy, Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, China
| | - Xinghong Jiang
- Key Laboratory of Pain Basic Research and Clinical Therapy, Department of Physiology and Neurobiology, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Jun JE, Kulhanek KR, Chen H, Chakraborty A, Roose JP. Alternative ZAP70-p38 signals prime a classical p38 pathway through LAT and SOS to support regulatory T cell differentiation. Sci Signal 2019; 12:12/591/eaao0736. [PMID: 31337738 DOI: 10.1126/scisignal.aao0736] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
T cell receptor (TCR) stimulation activates diverse kinase pathways, which include the mitogen-activated protein kinases (MAPKs) ERK and p38, the phosphoinositide 3-kinases (PI3Ks), and the kinase mTOR. Although TCR stimulation activates the p38 pathway through a "classical" MAPK cascade that is mediated by the adaptor protein LAT, it also stimulates an "alternative" pathway in which p38 is activated by the kinase ZAP70. Here, we used dual-parameter, phosphoflow cytometry and in silico computation to investigate how both classical and alternative p38 pathways contribute to T cell activation. We found that basal ZAP70 activation in resting T cell lines reduced the threshold ("primed") TCR-stimulated activation of the classical p38 pathway. Classical p38 signals were reduced after T cell-specific deletion of the guanine nucleotide exchange factors Sos1 and Sos2, which are essential LAT signalosome components. As a consequence of Sos1/2 deficiency, production of the cytokine IL-2 was impaired, differentiation into regulatory T cells was reduced, and the autoimmune disease EAE was exacerbated in mice. These data suggest that the classical and alternative p38 activation pathways exist to generate immune balance.
Collapse
Affiliation(s)
- Jesse E Jun
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kayla R Kulhanek
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hang Chen
- Departments of Chemical Engineering, Chemistry, and Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Arup Chakraborty
- Departments of Chemical Engineering, Chemistry, and Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Jeroen P Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
13
|
Yang W, Meng L, Chen K, Tian C, Peng B, Zhong L, Zhang C, Yang X, Zou J, Yang S, Li L. Preclinical pharmacodynamic evaluation of a new Src/FOSL1 inhibitor, LY-1816, in pancreatic ductal adenocarcinoma. Cancer Sci 2019; 110:1408-1419. [PMID: 30618127 PMCID: PMC6447837 DOI: 10.1111/cas.13929] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/10/2018] [Accepted: 12/25/2018] [Indexed: 02/05/2023] Open
Abstract
Despite tremendous efforts, the clinical prognosis of pancreatic ductal adenocarcinoma (PDAC) remains disappointing. There is an urgent need to develop more effective treatment strategies to improve the prognosis of patients with PDAC. In this study, we evaluate the anti‐PDAC effects of LY‐1816, a new multikinase inhibitor developed by us. In in vitro assays, LY‐1816 showed significant inhibitory effects on the proliferation, migration, and invasion of human PDAC cells, and induced PDAC cell apoptosis. Western blot analysis revealed that LY‐1816 markedly suppressed the Src signaling, and downregulated the expression of FOSL1; FOSL1 is an oncogene vulnerability in KRAS‐driven pancreatic cancer. In in vivo models of PDAC xenografts (Aspc‐1 and Bxpc‐3), LY‐1816 showed more potent antitumor activity than dasatinib and gemcitabine. Moreover, mice treated with LY‐1816 showed a much more significant survival advantage in a metastatic model of PDAC compared with those treated with vehicle, dasatinib, or gemcitabine. These results provide effective support for the subsequent clinical evaluation of LY‐1816 in the treatment of PDAC.
Collapse
Affiliation(s)
- Wei Yang
- Key Laboratory of Drug-Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Lingwei Meng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chenyu Tian
- Key Laboratory of Drug-Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Bing Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chunhui Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Zou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Linli Li
- Key Laboratory of Drug-Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Sawaya MR, Verma M, Balendiran V, Rath NP, Cascio D, Balendiran GK. Characterization of WY 14,643 and its Complex with Aldose Reductase. Sci Rep 2016; 6:34394. [PMID: 27721416 PMCID: PMC5056380 DOI: 10.1038/srep34394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 09/08/2016] [Indexed: 12/03/2022] Open
Abstract
The peroxisome proliferator, WY 14,643 exhibits a pure non-competitive inhibition pattern in the aldehyde reduction and in alcohol oxidation activities of human Aldose reductase (hAR). Fluorescence emission measurements of the equilibrium dissociation constants, Kd, of oxidized (hAR•NADP+) and reduced (hAR•NADPH) holoenzyme complexes display a 2-fold difference between them. Kd values for the dissociation of WY 14,643 from the oxidized (hAR•NADP+•WY 14,643) and reduced (hAR•NADPH•WY 14,643) ternary complexes are comparable to each other. The ternary complex structure of hAR•NADP+•WY 14,643 reveals the first structural evidence of a fibrate class drug binding to hAR. These observations demonstrate how fibrate molecules such as WY 14,643, besides being valued as agonists for PPAR, also inhibit hAR.
Collapse
Affiliation(s)
- Michael R. Sawaya
- UCLA-DOE, 611 Charles E. Young Drive East, 220 Boyer Hall, Los Angeles, CA 90095, USA
| | - Malkhey Verma
- Manchester Interdisciplinary Biocentre, 131 Princess Street, The University of Manchester, Manchester, M1 7DN, UK
| | - Vaishnavi Balendiran
- Department of Chemistry, WBSH 6017, Youngstown State University, One University Plaza, Youngstown, OH 44555, USA
| | - Nigam P. Rath
- Department of Chemistry and Biochemistry, University of Missouri-St. Louis, St. Louis, MO 63121, USA
| | - Duilio Cascio
- UCLA-DOE, 611 Charles E. Young Drive East, 220 Boyer Hall, Los Angeles, CA 90095, USA
| | - Ganesaratnam K. Balendiran
- Department of Chemistry, WBSH 6017, Youngstown State University, One University Plaza, Youngstown, OH 44555, USA
| |
Collapse
|
15
|
Yosef E, Katz A, Peleg Y, Mehlman T, Karlish SJD. Do Src Kinase and Caveolin Interact Directly with Na,K-ATPase? J Biol Chem 2016; 291:11736-50. [PMID: 27022017 DOI: 10.1074/jbc.m116.721084] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Indexed: 12/14/2022] Open
Abstract
Much evidence points to a role of Na,K-ATPase in ouabain-dependent signal transduction. Based on experiments with different cell lines and native tissue membranes, a current hypothesis postulates direct interactions between the Na,K-ATPase and Src kinase (non-receptor tyrosine kinase). Na,K-ATPase is proposed to bind Src kinase and inhibit its activity, whereas ouabain, the specific Na,K-ATPase inhibitor, binds and stabilizes the E2 conformation, thus exposing the Src kinase domain and its active site Tyr-418 for activation. Ouabain-dependent signaling is thought to be mediated within caveolae by a complex consisting of Na,K-ATPase, caveolin, and Src kinase. In the current work, we have looked for direct interactions utilizing purified recombinant Na,K-ATPase (human α1β1FXYD1 or porcine α1D369Nβ1FXYD1) and purified human Src kinase and human caveolin 1 or interactions between these proteins in native membrane vesicles isolated from rabbit kidney. By several independent criteria and techniques, no stable interactions were detected between Na,K-ATPase and purified Src kinase. Na,K-ATPase was found to be a substrate for Src kinase phosphorylation at Tyr-144. Clear evidence for a direct interaction between purified human Na,K-ATPase and human caveolin was obtained, albeit with a low molar stoichiometry (1:15-30 caveolin 1/Na,K-ATPase). In native renal membranes, a specific caveolin 14-5 oligomer (95 kDa) was found to be in direct interaction with Na,K-ATPase. We inferred that a small fraction of the renal Na,K-ATPase molecules is in a ∼1:1 complex with a caveolin 14-5 oligomer. Thus, overall, whereas a direct caveolin 1/Na,K-ATPase interaction is confirmed, the lack of direct Src kinase/Na,K-ATPase binding requires reassessment of the mechanism of ouabain-dependent signaling.
Collapse
Affiliation(s)
| | | | - Yoav Peleg
- the Israel Structural Proteomics Center, and
| | - Tevie Mehlman
- the Biological Services Department-Mass Spectrometry unit, Weizmann Institute of Science, Rehovot 7610001, Israel
| | | |
Collapse
|
16
|
Progesterone and Src family inhibitor PP1 synergistically inhibit cell migration and invasion of human basal phenotype breast cancer cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:426429. [PMID: 26075237 PMCID: PMC4449873 DOI: 10.1155/2015/426429] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 11/19/2014] [Indexed: 01/01/2023]
Abstract
Basal phenotype breast cancer is one of the most aggressive breast cancers that frequently metastasize to brain. The role of sex hormones and their receptors in development of this disease is largely unclear. We demonstrated that mPRα was expressed at a moderate level in a brain metastatic BPBC cell line MB231Br, which was derived from the parent mPRα undetectable MB231 cells. It functioned as an essential mediator for progesterone induced inhibitory effects on cell migration of MB231Br and, when coincubated with PP1, synergistically enhanced the progesterone's inhibitory effect on cell migration and invasion in vitro. Progesterone and PP1 cotreatment induced a cascade of molecular signaling events, such as dephosphorylation of FAK, downregulation of MMP9, VEGF, and KCNMA1 expressions. Our in vitro study demonstrated that mPRα was expressed and functioned as an essential mediator for progesterone induced inhibitory effects on cell migration and invasion in BPBC cells. This inhibitory effect was enhanced by PP1 via FAK dephosphorylation, MMP9, VEGF, and KCNMA1 downregulation mechanisms. Our study provides a new clue toward the development of novel promising agents and pathways for inhibiting nuclear hormonal receptor-negative and endocrine-resistant breast cancers.
Collapse
|
17
|
Pezzato C, Zaramella D, Martinelli M, Pieters G, Pagano MA, Prins LJ. Label-free fluorescence detection of kinase activity using a gold nanoparticle based indicator displacement assay. Org Biomol Chem 2014; 13:1198-203. [PMID: 25427977 DOI: 10.1039/c4ob02052a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A straightforward indicator-displacement assay (IDA) has been developed for the quantitative analysis of ATP→ADP conversion. The IDA relies on the use of gold nanoparticles passivated with a monolayer of thiols terminating with a 1,4,7-triazacyclononane (TACN)·Zn(2+) head group. The analytes ATP and ADP compete to a different extent with a fluorescent probe for binding to the monolayer surface. In the presence of ATP the fluorescent probe is free in solution, whereas in the presence of ADP the fluorescent probe is captured by the nanoparticles and its fluorescence is quenched. The linear response of the fluorescence signal towards different ratios of ATP : ADP permitted the detection of protein kinase activity simply by adding aliquots of the enzyme solution to the assay solution followed by measurement of the fluorescent intensity. The assay poses no restrictions on the target kinase nor does it require labeling of the kinase substrate. The assay was tested on the protein kinases PIM-1 and Src and validated through a direct comparison with the classical radiometric assay using the [γ-(32)P]-labeled ATP.
Collapse
Affiliation(s)
- Cristian Pezzato
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, 35131 Padova, Italy.
| | | | | | | | | | | |
Collapse
|
18
|
Frauenstein K, Tigges J, Soshilov AA, Kado S, Raab N, Fritsche E, Haendeler J, Denison MS, Vogel CFA, Haarmann-Stemmann T. Activation of the aryl hydrocarbon receptor by the widely used Src family kinase inhibitor 4-amino-5-(4-chlorophenyl)-7-(dimethylethyl)pyrazolo[3,4-d]pyrimidine (PP2). Arch Toxicol 2014; 89:1329-36. [PMID: 25082669 DOI: 10.1007/s00204-014-1321-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 07/21/2014] [Indexed: 01/16/2023]
Abstract
Small molecular weight protein kinase inhibitors are frequently used tools to unravel the complex network of cellular signal transduction under certain physiological and pathophysiological conditions. 4-amino-5-(4-chlorophenyl)-7-(dimethylethyl)pyrazolo[3,4-d]pyrimidine (PP2) is a widely used compound to block the activity of Src family kinases, the major group of non-receptor tyrosine kinases, which trigger multiple cellular signaling pathways. Here, we show that PP2 induces cytochrome P450 1A1 mRNA expression and enzyme activity in a dose-dependent manner in human HepG2 hepatoma cells and NCTC 2544 keratinocytes. By means of reporter gene assays, RNA interference, electrophoretic mobility shift assay, and competitive ligand-binding assay, we further demonstrate that PP2 is a ligand for the aryl hydrocarbon receptor (AHR), an intracellular chemosensor that regulates xenobiotic metabolism, environmental stress responses, and immune functions. Upon ligand-dependent activation, the AHR translocates into the nucleus and dimerizes with the AHR nuclear translocator (ARNT) to modulate the expression of its target genes. In addition, AHR activation is frequently accompanied by an activation of the tyrosine kinase c-Src, resulting in stimulation of cell-surface receptors and downstream signal transduction. As PP2 activates the AHR/ARNT pathway by simultaneously blocking c-Src-mediated alternative signaling routes, this compound may be a suitable tool to study the contribution of the different AHR-dependent signaling pathways to biological processes and adverse outcomes. On the other hand, the unexpected property of PP2 to stimulate AHR/ARNT signaling should be carefully taken into account in future investigations in order to avoid a false interpretation of experimental results and molecular interrelations.
Collapse
Affiliation(s)
- Katrin Frauenstein
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Irtegun S, Wood R, Lackovic K, Schweiggert J, Ramdzan YM, Huang DCS, Mulhern TD, Hatters DM. A biosensor of SRC family kinase conformation by exposable tetracysteine useful for cell-based screening. ACS Chem Biol 2014; 9:1426-31. [PMID: 24828008 DOI: 10.1021/cb500242q] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We developed a new approach to distinguish distinct protein conformations in live cells. The method, exposable tetracysteine (XTC), involved placing an engineered tetracysteine motif into a target protein that has conditional access to biarsenical dye binding by conformational state. XTC was used to distinguish open and closed regulatory conformations of Src family kinases. Substituting just four residues with cysteines in the conserved SH2 domain of three Src-family kinases (c-Src, Lck, Lyn) enabled open and closed conformations to be monitored on the basis of binding differences to biarsenical dyes FlAsH or ReAsH. Fusion of the kinases with a fluorescent protein tracked the kinase presence, and the XTC approach enabled simultaneous assessment of regulatory state. The c-Src XTC biosensor was applied in a boutique screen of kinase inhibitors, which revealed six compounds to induce conformational closure. The XTC approach demonstrates new potential for assays targeting conformational changes in key proteins in disease and biology.
Collapse
Affiliation(s)
| | | | - Kurt Lackovic
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | | | | | - David C. S. Huang
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | | | | |
Collapse
|
20
|
Holbrook BC, Yammani RD, Blevins LK, Alexander-Miller MA. In vivo modulation of avidity in highly sensitive CD8(+) effector T cells following viral infection. Viral Immunol 2013; 26:302-13. [PMID: 23971914 DOI: 10.1089/vim.2013.0042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Numerous studies have demonstrated a critical role for T cell avidity in predicting in vivo efficacy. Even though the measurement of avidity is now a routine assessment for the analysis of effector and memory T cell populations, our understanding of how this property is controlled in vivo at both the population and individual cell levels is limited. Our previous studies have identified high avidity as a property of the initial effector population generated in mice following respiratory virus infection. As the response progresses, lower avidity cells appear in the effector pool. The studies described here investigate the mechanistic basis of this in vivo regulation of avidity. We present data supporting in vivo avidity modulation within the early high avidity responders that results in a population of lower avidity effector cells. Changes in avidity were correlated with decreased lck expression and increased sensitivity to lck inhibitors in effector cells present at late versus early times postinfection. The possibility of tuning within select individual effectors is a previously unappreciated mechanism for the control of avidity in vivo.
Collapse
Affiliation(s)
- Beth C Holbrook
- Department of Microbiology and Immunology, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | | | | | | |
Collapse
|
21
|
Xie M, You S, Chen Q, Chen X, Hu C. Progesterone inhibits the migration and invasion of A549 lung cancer cells through membrane progesterone receptor α-mediated mechanisms. Oncol Rep 2013; 29:1873-80. [PMID: 23467643 DOI: 10.3892/or.2013.2336] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 02/18/2013] [Indexed: 11/06/2022] Open
Abstract
Lung cancer is the leading cause of cancer morbidity and mortality in the world. The incidence of lung cancer, particularly lung adenocarcinoma, is increasing in women compared to men. The role of sex hormones in the development of lung cancer has attracted substantial interest, but remains largely unknown. In this study, we demonstrated that membrane progesterone receptor α (mPRα) was expressed in a lung adenocarcinoma cell line, A549, and was located on the cell membrane. In additional experiments, we found that mPRα functioned as an essential mediator for progesterone (P4)-induced inhibitory effects on cell migration and invasion of A549 cells. Furthermore, PP1 (an Src pathway inhibitor), when co-incubated with P4, synchronously enhanced the inhibitory effects of P4 on cell migration and invasion. To explore the mechanisms of inhibition, we found that P4 and PP1 induced a cascade of molecular signaling events, such as dephosphorylation of focal adhesion kinase (FAK) and downregulation of matrix metalloproteinase 9 (MMP-9). Our study provides a mechanistic view on the effects of P4 through mPRα→Src/FAK relevant pathways in human lung adenocarcinoma cells and may aid in the development of novel therapeutic tools for the treatment of lung cancer.
Collapse
Affiliation(s)
- Mingxuan Xie
- Department of Gerontology and Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | | | | | | | | |
Collapse
|
22
|
Seiler C, Davuluri G, Abrams J, Byfield FJ, Janmey PA, Pack M. Smooth muscle tension induces invasive remodeling of the zebrafish intestine. PLoS Biol 2012; 10:e1001386. [PMID: 22973180 PMCID: PMC3433428 DOI: 10.1371/journal.pbio.1001386] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 07/26/2012] [Indexed: 12/12/2022] Open
Abstract
The signals that initiate cell invasion are not well understood, but there is increasing evidence that extracellular physical signals play an important role. Here we show that epithelial cell invasion in the intestine of zebrafish meltdown (mlt) mutants arises in response to unregulated contractile tone in the surrounding smooth muscle cell layer. Physical signaling in mlt drives formation of membrane protrusions within the epithelium that resemble invadopodia, matrix-degrading protrusions present in invasive cancer cells. Knockdown of Tks5, a Src substrate that is required for invadopodia formation in mammalian cells blocked formation of the protrusions and rescued invasion in mlt. Activation of Src-signaling induced invadopodia-like protrusions in wild type epithelial cells, however the cells did not migrate into the tissue stroma, thus indicating that the protrusions were required but not sufficient for invasion in this in vivo model. Transcriptional profiling experiments showed that genes responsive to reactive oxygen species (ROS) were upregulated in mlt larvae. ROS generators induced invadopodia-like protrusions and invasion in heterozygous mlt larvae but had no effect in wild type larvae. Co-activation of oncogenic Ras and Wnt signaling enhanced the responsiveness of mlt heterozygotes to the ROS generators. These findings present the first direct evidence that invadopodia play a role in tissue cell invasion in vivo. In addition, they identify an inducible physical signaling pathway sensitive to redox and oncogenic signaling that can drive this process.
Collapse
Affiliation(s)
- Christoph Seiler
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Gangarao Davuluri
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Joshua Abrams
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Fitzroy J. Byfield
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Paul A. Janmey
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael Pack
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
23
|
Feng S, Pflueger M, Lin SX, Groveman BR, Su J, Yu XM. Regulation of voltage-gated sodium current by endogenous Src family kinases in cochlear spiral ganglion neurons in culture. Pflugers Arch 2012; 463:571-84. [PMID: 22297656 DOI: 10.1007/s00424-012-1072-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2011] [Revised: 12/09/2011] [Accepted: 01/02/2012] [Indexed: 01/28/2023]
Abstract
Voltage-gated sodium (Na+) and potassium (K+)channels have been found to be regulated by Src family kinases(SFKs).However, how these channels are regulated by SFKs in cochlear spiral ganglion neurons (SGNs) remains unknown.Here, we report that altering the activity of endogenous SFKs modulated voltage-gated Na+, but not K+, currents recorded in embryonic SGNs in culture. Voltage-gated Na+ current was suppressed by inhibition of endogenous SFKs or just Src and potentiated by the activation of these enzymes. Detailed investigations showed that under basal conditions, SFK inhibitor application did not significantly affect the voltage-dependent activation, but shifted the steady-state inactivation curves of Na+ currents and delayed the recovery of Na+ currents from inactivation. Application of Src specific inhibitor, Src40–58,not only shifted the inactivation curve but also delayed the recovery of Na+ currents and moved the voltage-dependent activation curve towards the left. The pre-inhibition of SFKs occluded all the effects induced by Src40–58 application, except the left shift of the activation curve. The activation of SFKs did not change either steady-state inactivation or recovery of Na+ currents, but caused the left shift of the activation curve.SFK inhibitor application effectively prevented all the effects induced by SFK activation, suggesting that both the voltage-dependent activation and steady-state inactivation of Na+ current are subjects of SFK regulation. The different effects induced by activation versus inhibition of SFKs implied that under basal conditions, endogenously active and inactive SFKs might be differentially involved in the regulation of voltage-gated Na+ channels in SGNs.
Collapse
Affiliation(s)
- Shuang Feng
- Department of Otolaryngology—Head and Neck Surgery, First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | | | | | | | | | | |
Collapse
|
24
|
Ray RM, Li C, Bhattacharya S, Naren AP, Johnson LR. Spermine, a molecular switch regulating EGFR, integrin β3, Src, and FAK scaffolding. Cell Signal 2012; 24:931-942. [PMID: 22227249 PMCID: PMC3334284 DOI: 10.1016/j.cellsig.2011.12.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 12/19/2011] [Indexed: 12/21/2022]
Abstract
Intracellular polyamine levels are highly regulated by the activity of ornithine decarboxylase (ODC), which catalyzes the first rate-limiting reaction in polyamine biosynthesis, producing putrescine, which is subsequently converted to spermidine and spermine. We have shown that polyamines regulate proliferation, migration, and apoptosis in intestinal epithelial cells. Polyamines regulate key signaling events at the level of the EGFR and Src. However, the precise mechanism of action of polyamines is unknown. In the present study, we demonstrate that ODC localizes in lamellipodia and in adhesion plaques during cell spreading. Spermine regulates EGF-induced migration by modulating the interaction of the EGFR with Src. The EGFR interacted with integrin β3, Src, and focal adhesion kinase (FAK). Active Src (pY418-Src) localized with FAK during spreading and migration. Spermine prevented EGF-induced binding of the EGFR with integrin β3, Src, and FAK. Activation of Src and FAK was necessary for EGF-induced migration in HEK293 cells. EGFR-mediated Src activation in live HEK293 cells using a FRET based Src reporter showed that polyamine depletion significantly increased Src kinase activity. In vitro binding studies showed that spermine directly binds Src, and preferentially interacts with the SH2 domain of Src. The physical interaction between Src and the EGFR was severely attenuated by spermine. Therefore, spermine acts as a molecular switch in regulating EGFR-Src coupling both physically and functionally. Upon activation of the EGFR, integrin β3, FAK and Src are recruited to EGFR leading to the trans-activation of both the EGFR and Src and to the Src-mediated phosphorylation of FAK. The activation of FAK induced Rho-GTPases and subsequently migration. This is the first study to define mechanistically how polyamines modulate Src function at the molecular level.
Collapse
Affiliation(s)
- Ramesh M Ray
- Department of Physiology, The University of Tennessee Health Science Center, 894 Union Avenue, Memphis, TN 38163, USA.
| | | | | | | | | |
Collapse
|
25
|
Giannatselis H, Calder M, Watson AJ. Ouabain stimulates a Na+/K+-ATPase-mediated SFK-activated signalling pathway that regulates tight junction function in the mouse blastocyst. PLoS One 2011; 6:e23704. [PMID: 21901128 PMCID: PMC3162003 DOI: 10.1371/journal.pone.0023704] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 07/23/2011] [Indexed: 01/22/2023] Open
Abstract
The Na(+)/K(+)-ATPase plays a pivotal role during preimplantation development; it establishes a trans-epithelial ionic gradient that facilitates the formation of the fluid-filled blastocyst cavity, crucial for implantation and successful pregnancy. The Na(+)/K(+)-ATPase is also implicated in regulating tight junctions and cardiotonic steroid (CTS)-induced signal transduction via SRC. We investigated the expression of SRC family kinase (SFK) members, Src and Yes, during preimplantation development and determined whether SFK activity is required for blastocyst formation. Embryos were collected following super-ovulation of CD1 or MF1 female mice. RT-PCR was used to detect SFK mRNAs encoding Src and Yes throughout preimplantation development. SRC and YES protein were localized throughout preimplantation development. Treatment of mouse morulae with the SFK inhibitors PP2 and SU6656 for 18 hours resulted in a reversible blockade of progression to the blastocyst stage. Blastocysts treated with 10(-3) M ouabain for 2 or 10 minutes and immediately immunostained for phosphorylation at SRC tyr418 displayed reduced phosphorylation while in contrast blastocysts treated with 10(-4) M displayed increased tyr418 fluorescence. SFK inhibition increased and SFK activation reduced trophectoderm tight junction permeability in blastocysts. The results demonstrate that SFKs are expressed during preimplantation development and that SFK activity is required for blastocyst formation and is an important mediator of trophectoderm tight junction permeability.
Collapse
Affiliation(s)
- Holly Giannatselis
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - Michele Calder
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - Andrew J. Watson
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
26
|
Yokdang N, Tellez JD, Tian H, Norvell J, Barsky SH, Valencik M, Buxton ILO. A role for nucleotides in support of breast cancer angiogenesis: heterologous receptor signalling. Br J Cancer 2011; 104:1628-40. [PMID: 21505453 PMCID: PMC3101911 DOI: 10.1038/bjc.2011.134] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background: Human breast carcinoma cells secrete an adenosine 5′-diphosphate transphosphorylase (sNDPK) known to induce endothelial cell tubulogenesis in a P2Y receptor-dependent manner. We examined sNDPK secretion and its effects on human endothelial cells. Methods: Nucleoside diphosphate kinase (NDPK) secretion was measured by western blot and enzyme-linked immunosorbent assay, while transphosphorylase activity was measured using the luciferin-luciferase ATP assay. Activation of MAPK was determined by western blot analysis, immunofluorescence and endothelial cell proliferation and migration. Results: A panel of breast cancer cell lines with origin as ductal carcinoma, adenocarcinoma or medullary carcinoma, secrete sNDPK-A/B. Addition of purified NDPK-B to endothelial cultures activated VEGFR-2 and Erk1/2, both of which were blocked by inhibitors of NDPK and P2Y receptors. Activation of VEGFR-2 and ErK1/2 by 2-methylthio-ATP (2MeS-ATP) was blocked by pretreatment with the P2Y1-specific antagonist MRS2179, the proto-oncogene non-receptor tyrosine kinase (Src) inhibitor PP2 or the VEGFR-2 antagonist SU1498. Nucleoside diphosphate kinase-B stimulates cell growth and migration in a concentration-dependent manner comparable to the effect of vascular endothelial growth factor. Treatment of endothelial cells with either NDPK-B or 2MeS-ATP induced migration, blocked by P2Y1, Src or VEGFR-2 antagonists. Conclusion: sNDPK supports angiogenesis. Understanding the mechanism of action of sNDPK and P2Y1 nucleotide signalling in metastasis and angiogenesis represent new therapeutic targets for anti-angiogenic therapies to benefit patients.
Collapse
Affiliation(s)
- N Yokdang
- Department of Pharmacology, Centre for Molecular Medicine, University of Nevada School of Medicine, Mail Stop 573, Reno, NV 89557, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Eathiraj S, Palma R, Hirschi M, Volckova E, Nakuci E, Castro J, Chen CR, Chan TCK, France DS, Ashwell MA. A novel mode of protein kinase inhibition exploiting hydrophobic motifs of autoinhibited kinases: discovery of ATP-independent inhibitors of fibroblast growth factor receptor. J Biol Chem 2011; 286:20677-87. [PMID: 21454610 DOI: 10.1074/jbc.m110.213736] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Protein kinase inhibitors with enhanced selectivity can be designed by optimizing binding interactions with less conserved inactive conformations because such inhibitors will be less likely to compete with ATP for binding and therefore may be less impacted by high intracellular concentrations of ATP. Analysis of the ATP-binding cleft in a number of inactive protein kinases, particularly in the autoinhibited conformation, led to the identification of a previously undisclosed non-polar region in this cleft. This ATP-incompatible hydrophobic region is distinct from the previously characterized hydrophobic allosteric back pocket, as well as the main pocket. Generalized hypothetical models of inactive kinases were constructed and, for the work described here, we selected the fibroblast growth factor receptor (FGFR) tyrosine kinase family as a case study. Initial optimization of a FGFR2 inhibitor identified from a library of commercial compounds was guided using structural information from the model. We describe the inhibitory characteristics of this compound in biophysical, biochemical, and cell-based assays, and have characterized the binding mode using x-ray crystallographic studies. The results demonstrate, as expected, that these inhibitors prevent activation of the autoinhibited conformation, retain full inhibitory potency in the presence of physiological concentrations of ATP, and have favorable inhibitory activity in cancer cells. Given the widespread regulation of kinases by autoinhibitory mechanisms, the approach described herein provides a new paradigm for the discovery of inhibitors by targeting inactive conformations of protein kinases.
Collapse
|
28
|
Fang XQ, Xu J, Feng S, Groveman BR, Lin SX, Yu XM. The NMDA receptor NR1 subunit is critically involved in the regulation of NMDA receptor activity by C-terminal Src kinase (Csk). Neurochem Res 2011; 36:319-26. [PMID: 21113815 PMCID: PMC3032389 DOI: 10.1007/s11064-010-0330-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 11/15/2010] [Indexed: 01/25/2023]
Abstract
Previous studies have shown that Csk plays critical roles in the regulation of neural development, differentiation and glutamate-mediated synaptic plasticity. It has been found that Csk associates with the NR2A and 2B subunits of N-methyl-D-aspartate receptors (NMDARs) in a Src activity-dependent manner and serves as an intrinsic mechanism to provide a "brake" on the induction of long-term synaptic potentiation (LTP) mediated by NMDARs. In contrast to the NR2A and 2B subunits, no apparent tyrosine phosphorylation is found in the NR1 subunit of NMDARs. Here, we report that Csk can also associate with the NR1 subunit in a Src activity-dependent manner. The truncation of the NR1 subunit C-tail which contains only one tyrosine (Y837) significantly reduced the Csk association with the NR1-1a/NR2A receptor complex. Furthermore, we found that either the truncation of NR2A C-tail at aa 857 or the mutation of Y837 in the NR1-1a subunit to phenylalanine blocked the inhibition of NR1-1a/NR2A receptors induced by intracellular application of Csk. Thus, both the NR1 and NR2 subunits are required for the regulation of NMDAR activity by Csk.
Collapse
Affiliation(s)
- Xiao-Qian Fang
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL 32306-4300, USA
| | - Jindong Xu
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Shuang Feng
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL 32306-4300, USA. Department of Otolaryngology-Head and Neck Surgery, Guangxi Medical University, 530021 Nanning, People’s Republic of China
| | - Bradley R. Groveman
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL 32306-4300, USA
| | - Shuang-Xiu Lin
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL 32306-4300, USA
| | - Xian-Min Yu
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahassee, FL 32306-4300, USA. Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| |
Collapse
|
29
|
Marin V, Groveman BR, Qiao H, Xu J, Ali MK, Fang XQ, Lin S, Rizkallah R, Hurt MH, Bienkiewicz EA, Yu XM. Characterization of neuronal Src kinase purified from a bacterial expression system. Protein Expr Purif 2010; 74:289-97. [PMID: 20558296 PMCID: PMC2952679 DOI: 10.1016/j.pep.2010.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 06/08/2010] [Accepted: 06/08/2010] [Indexed: 11/17/2022]
Abstract
Neuronal Src (n-Src) is an alternative isoform of Src kinase containing a 6-amino acid insert in the SH3 domain that is highly expressed in neurons of the central nervous system (CNS). To investigate the function of n-Src, wild-type n-Src, constitutively active n-Src in which the C-tail tyrosine 535 was mutated to phenylalanine (n-Src/Y535F) and inactive n-Src in which the lysine 303 was mutated to arginine in addition to the mutation of Y535F (n-Src/K303R/Y535F), were expressed and purified from Escherichia coli BL21(DE3) cells. We found that all three types of n-Src constructs expressed at very high yields (∼500 mg/L) at 37°C, but formed inclusion bodies. In the presence of 8M urea these proteins could be solubilized, purified under denaturing conditions, and subsequently refolded in the presence of arginine (0.5M). These Src proteins were enzymatically active except for the n-Src/K303R/Y535F mutant. n-Src proteins expressed at 18°C were soluble, albeit at lower yields (∼10-20 mg/L). The lowest yields were for n-Src/Y535F (∼10 mg/L) and the highest for n-Src/K303R/Y535F (∼20 mg/L). We characterized the purified n-Src proteins expressed at 18°C. We found that altering n-Src enzyme activity either pharmacologically (e.g., application of ATP or a Src inhibitor) or genetically (mutation of Y535 or K303) was consistently associated with changes in n-Src stability: an increase in n-Src activity was coupled with a decrease in n-Src stability and vice versa. These findings, therefore, indicate that n-Src activity and stability are interdependent. Finally, the successful production of functionally active n-Src in this study indicates that the bacterial expression system may be a useful protein source in future investigations of n-Src regulation and function.
Collapse
Affiliation(s)
- Vedrana Marin
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee FL 32306-4300, USA
| | - Bradley R. Groveman
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee FL 32306-4300, USA
| | - Haifa Qiao
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee FL 32306-4300, USA
| | - Jindong Xu
- Faculty of Dentistry, University of Toronto, Ontario M5G 1G6, Canada
| | - Mohammad K. Ali
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee FL 32306-4300, USA
| | - Xiao-Qian Fang
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee FL 32306-4300, USA
| | - Shuangxiu Lin
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee FL 32306-4300, USA
| | - Raed Rizkallah
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee FL 32306-4300, USA
| | - Myra H. Hurt
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee FL 32306-4300, USA
| | - Ewa A. Bienkiewicz
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee FL 32306-4300, USA
| | - Xian-Min Yu
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee FL 32306-4300, USA
- Faculty of Dentistry, University of Toronto, Ontario M5G 1G6, Canada
| |
Collapse
|
30
|
Herraiz C, Journé F, Abdel-Malek Z, Ghanem G, Jiménez-Cervantes C, García-Borrón JC. Signaling from the human melanocortin 1 receptor to ERK1 and ERK2 mitogen-activated protein kinases involves transactivation of cKIT. Mol Endocrinol 2010; 25:138-56. [PMID: 21084381 DOI: 10.1210/me.2010-0217] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Melanocortin 1 receptor (MC1R), a Gs protein-coupled receptor expressed in melanocytes, is a major determinant of skin pigmentation, phototype and cancer risk. Upon stimulation by αMSH, MC1R triggers the cAMP and ERK1/ERK2 MAPK pathways. In mouse melanocytes, ERK activation by αMSH binding to Mc1r depends on cAMP, and melanocytes are considered a paradigm for cAMP-dependent ERK activation. However, human MC1R variants associated with red hair, fair skin [red hair color (RHC) phenotype], and increased skin cancer risk display reduced cAMP signaling but activate ERKs as efficiently as wild type in heterologous cells, suggesting independent signaling to ERKs and cAMP in human melanocytes. We show that MC1R signaling activated the ERK pathway in normal human melanocytes and melanoma cells expressing physiological levels of endogenous RHC variants. ERK activation was comparable for wild-type and mutant MC1R and was independent on cAMP because it was neither triggered by stimulation of cAMP synthesis with forskolin nor blocked by the adenylyl cyclase inhibitor 2',5'-dideoxyadenosine. Stimulation of MC1R with αMSH did not lead to protein kinase C activation and ERK activation was unaffected by protein kinase C inhibitors. Conversely, pharmacological interference, small interfering RNA studies, expression profiles, and functional reconstitution experiments showed that αMSH-induced ERK activation resulted from Src tyrosine kinase-mediated transactivation of the stem cell factor receptor, a receptor tyrosine kinase essential for proliferation, differentiation, and survival of melanocyte precursors, thus demonstrating a functional link between the stem cell factor receptor and MC1R. Moreover, this transactivation phenomenon is unique because it is unaffected by natural mutations impairing canonical MC1R signaling through the cAMP pathway.
Collapse
Affiliation(s)
- Cecilia Herraiz
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia, Campus de Espinardo, 30100 Espinardo, Murcia, Spain
| | | | | | | | | | | |
Collapse
|
31
|
Kong L, Deng Z, Shen H, Zhang Y. Src family kinase inhibitor PP2 efficiently inhibits cervical cancer cell proliferation through down-regulating phospho-Src-Y416 and phospho-EGFR-Y1173. Mol Cell Biochem 2010; 348:11-9. [PMID: 21052789 DOI: 10.1007/s11010-010-0632-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Accepted: 10/18/2010] [Indexed: 11/26/2022]
Abstract
Tyrosine (Y) kinases inhibitors have been approved for targeted treatment of cancer. However, their clinical use is limited to some cancers and the mechanism of their action remains unclear. Previous study has indicated that PP2, a selective inhibitor of the Src family of non-receptor tyrosine kinases (nRTK), efficiently repressed cervical cancer growth in vitro and in vivo. In this regard, our aims are to explore the mechanism of PP2 on cervical cancer cell growth inhibition by investigating the suppressive divergence among PP1, PP2, and a negative control compound PP3. MTT results showed that three compounds had different inhibitory effects on proliferation of two cervical cancer cells, HeLa and SiHa, and PP2 was most efficient in a time- and dose-dependent manner. Moreover, we found 10 μM PP2 down-regulated pSrc-Y416 (P < 0.05), pEGFR-Y845 (P < 0.05), and -Y1173 (P < 0.05) expression levels, while 10 μM PP1 down-regulated pSrc-Y416 (P < 0.05) and pEGFR-Y845 (P < 0.05), but not pEGFR-Y1173; 10 μM PP3 down-regulated only pEGFR-Y1173 (P < 0.05). PP2 could modulate cell cycle arrest by up-regulating p21(Cip1) and p27(Kip1) in both HeLa and SiHa cells and down-regulating expression of cyclin A, and cyclin dependent kinase-2, -4 (Cdk-2, -4) in HeLa and of cyclin B and Cdk-2 in SiHa. Our results indicate that Src pathway and EGFR pathway play different roles in the proliferation of cervical cancer cells and PP2 efficiently reduces cervical cancer cell proliferation by reduction of both Src and EGFR activity.
Collapse
Affiliation(s)
- Lu Kong
- Department of Biochemistry and Molecular Biology, Cancer Institute, Capital Medical University, Beijing, China
| | | | | | | |
Collapse
|
32
|
Abstract
Classical enzymology has been used for generations to understand the interactions of inhibitors with their enzyme targets. Enzymology tools enabled prediction of the biological impact of inhibitors as well as the development of novel, more potent, ones. Experiments designed to examine the competition between the tested inhibitor and the enzyme substrate(s) are the tool of choice to identify inhibitors that bind in the active site. Competition between an inhibitor and a substrate is considered a strong evidence for binding of the inhibitor in the active site, while the lack of competition suggests binding to an alternative site. Nevertheless, exceptions to this notion do exist. Active site-binding inhibitors can display non-competitive inhibition patterns. This unusual behavior has been observed with enzymes utilizing an exosite for substrate binding, isomechanism enzymes, enzymes with multiple substrates and/or products and two-step binding inhibitors. In many of these cases, the mechanisms underlying the lack of competition between the substrate and the inhibitor are well understood. Tools like alternative substrates, testing the enzyme reaction in the reverse direction and monitoring inhibition time dependence can be applied to enable distinction between 'badly behaving' active site binders and true exosite inhibitors.
Collapse
Affiliation(s)
- Yuval Blat
- Department of Mechanistic Biochemistry, Bristol-Myers Squibb Company, Rt. 206 and Provinceline Rd., Princeton, NJ 08543, USA.
| |
Collapse
|
33
|
Amyloid precursor protein mediates a tyrosine kinase-dependent activation response in endothelial cells. J Neurosci 2009; 29:14451-62. [PMID: 19923279 DOI: 10.1523/jneurosci.3107-09.2009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Amyloid precursor protein (APP) is a ubiquitously expressed type 1 integral membrane protein. It has the ability to bind numerous extracellular matrix components and propagate signaling responses via its cytoplasmic phospho-tyrosine, (682)YENPTY(687), binding motif. We recently demonstrated increased protein levels of APP, phosphorylated APP (Tyr682), and beta-amyloid (Abeta) in brain vasculature of atherosclerotic and Alzheimer's disease (AD) tissue colocalizing primarily within the endothelial layer. This study demonstrates similar APP changes in peripheral vasculature from human and mouse apoE(-/-) aorta, suggesting that APP-related changes are not restricted to brain vasculature. Therefore, primary mouse aortic endothelial cells and human umbilical vein endothelial cells were used as a model system to examine the function of APP in endothelial cells. APP multimerization with an anti-N-terminal APP antibody, 22C11, to simulate ligand binding stimulated an Src kinase family-dependent increase in protein phospho-tyrosine levels, APP phosphorylation, and Abeta secretion. Furthermore, APP multimerization stimulated increased protein levels of the proinflammatory proteins, cyclooxygenase-2 and vascular cell adhesion molecule-1 also in an Src kinase family-dependent manner. Endothelial APP was also involved in mediating monocytic cell adhesion. Collectively, these data demonstrate that endothelial APP regulates immune cell adhesion and stimulates a tyrosine kinase-dependent response driving acquisition of a reactive endothelial phenotype. These APP-mediated events may serve as therapeutic targets for intervention in progressive vascular changes common to cerebrovascular disease and AD.
Collapse
|
34
|
Han A, Hosokawa K, Maeda M. Phosphate-affinity electrophoresis on a microchip for determination of protein kinase activity. Electrophoresis 2009; 30:3507-13. [DOI: 10.1002/elps.200900142] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
35
|
Chaturvedi LS, Saad SA, Bakshi N, Marsh HM, Basson MD. Strain matrix-dependently dissociates gut epithelial spreading and motility. J Surg Res 2009; 156:217-23. [PMID: 19691992 PMCID: PMC2749895 DOI: 10.1016/j.jss.2009.03.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 03/18/2009] [Accepted: 03/22/2009] [Indexed: 01/26/2023]
Abstract
BACKGROUND Repetitive deformation enhances intestinal epithelial migration across tissue fibronectin (tFN) via Src but inhibits migration across collagen. Since cell spreading generally precedes motility, we compared the effects of cyclic strain on Caco-2 spreading and migration on tFN, collagen-I, and plasma fibronectin (pFN), and investigated the role of Src in deformation-influenced spreading and migration. MATERIALS AND METHODS Human Caco-2 intestinal epithelial cells on tFN, collagen-I or pFN were subjected to an average 10% strain at 10 cycles/min for 2 h. Src was inhibited with 10muM PP2 or Src was reduced with siRNA. Parallel studies assessed deformation effects on monolayer wound closure. RESULTS Deformation, Src-inhibition or reduction each inhibited spreading on tFN but Src-inhibition or reduction prevented further inhibition of spreading by deformation without preventing further inhibition of motility. Deformation did not alter spreading on collagen-I or pFN, but inhibited wound closure. CONCLUSIONS Although cell spreading generally precedes and parallels motility, repetitive deformation regulates motility independently of spreading. Since deformation activates Src, the ability of Src blockade to mimic strain-associated inhibition of spreading on tFN suggests that this effect occurs by a separate mechanism that may also require basal Src activity. Further delineation of the mechanisms by which strain disparately modulates spreading and motility may permit acceleration of mucosal healing by targeted interventions to separately promote spreading and epithelial motility.
Collapse
Affiliation(s)
- Lakshmi S. Chaturvedi
- Departments of Surgery, John D. Dingell VA Medical Center
- Departments of Surgery, Wayne State University
| | - Samira A. Saad
- Departments of Surgery, John D. Dingell VA Medical Center
- Departments of Surgery, Wayne State University
| | - Neil Bakshi
- Departments of Surgery, John D. Dingell VA Medical Center
- Departments of Surgery, Wayne State University
| | - Harold M. Marsh
- Departments of Surgery, John D. Dingell VA Medical Center
- Departments of Surgery, Wayne State University
| | - Marc D. Basson
- Departments of Surgery, Michigan State University
- Departments of Surgery, John D. Dingell VA Medical Center
| |
Collapse
|
36
|
Cyclic stretch downregulates arterial vascular connexin43 protein expression: an ex vivo study. Biomech Model Mechanobiol 2009; 9:203-11. [DOI: 10.1007/s10237-009-0171-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Accepted: 08/10/2009] [Indexed: 11/26/2022]
|
37
|
Raué HP, Slifka MK. CD8+ T cell immunodominance shifts during the early stages of acute LCMV infection independently from functional avidity maturation. Virology 2009; 390:197-204. [PMID: 19539966 PMCID: PMC2728041 DOI: 10.1016/j.virol.2009.05.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 05/14/2009] [Accepted: 05/16/2009] [Indexed: 02/01/2023]
Abstract
Virus-specific T cell responses are often directed to a small subset of possible epitopes and their relative magnitude defines their hierarchy. We determined the size and functional avidity of 4 representative peptide-specific CD8(+) T cell populations in C57BL/6 mice at different time points after lymphocytic choriomeningitis virus (LCMV) infection. We found that the frequency of different peptide-specific T cell populations in the spleen changed independently over the first 8 days after infection. These changes were not associated with a larger or more rapid increase in functional avidity and yet still resulted in a shift in the final immunodominance hierarchy. Thus, the immunodominance observed at the peak of an antiviral T cell response is not necessarily determined by the initial size or rate of functional avidity maturation of peptide-specific T cell populations.
Collapse
Affiliation(s)
- Hans-Peter Raué
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Mark K. Slifka
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| |
Collapse
|
38
|
Zuckerman ST, Brown JF, Kao WJ. Identification of regulatory Hck and PAI-2 proteins in the monocyte response to PEG-containing matrices. Biomaterials 2009; 30:3825-33. [PMID: 19443025 PMCID: PMC2774525 DOI: 10.1016/j.biomaterials.2009.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 04/13/2009] [Indexed: 11/29/2022]
Abstract
Mass spectrometry is a powerful proteomic tool enabling researchers to survey the global proteome of a cell. This technique has only recently been employed to investigate cell-material interactions. We had previously identified material scarcity and limited adherent cells as challenges facing mass spectrometric analysis of cell-material interactions. U937 adherent to tissue culture poly(styrene) was used as a model system for identifying proteins expressed by adherent monocytes and analyzed by HPLC coupled offline to MALDI-ToF/ToF (LC-MALDI). We identified 645 proteins from two cation fractions of crude U937 monocyte cell lysate. Forty three proteins of interest from the 645 were chosen based on literature searches for relevance to monocyte-material inflammation and wound healing. Proteins such as 40S ribosomal protein S19 and tyrosyl tRNA synthetase highlight the ability of LC-MALDI to identify proteins relevant to monocyte-material interactions that are currently unexplored. We used PEG-based semi-interpenetrating polymer networks and PEG-only hydrogels to investigate surface dependent effects on the Src family kinase Hck and plasminogen activator inhibitor-2 (PAI-2) using the pyrazolo pyrimidine small molecule inhibitor PP2 and exogenous urokinase plasminogen activator addition, respectively. Hck is well researched in cell adhesion while PAI-2 is virtually unknown in cell-material interactions. U937 on TCPS and PEG-only hydrogels secreted similar levels of inflammatory cytokines and gelatinase MMP-9. MCP-1 secretion from monocytes on PEG-only hydrogels was Hck independent in contrast to Hck-dependent MCP-1 secretion in U937 on TCPS. Overall, U937 adherent to sIPNs secrete low levels of soluble gelatinase MMP-9, IL-1beta, TNF-alpha, IL-6, and MCP-1 independent of Hck and PAI-2. This work demonstrates significant changes in surface dependent expression of proteins from monocytes adherent to PEG-based materials compared to TCPS.
Collapse
Affiliation(s)
- Sean T. Zuckerman
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, 53705 USA
- School of Pharmacy, University of Wisconsin-Madison, WI, 53705 USA
| | - James F. Brown
- Biotechnology Center, University of Wisconsin-Madison, WI, 53705 USA
| | - Weiyuan John Kao
- Department of Biomedical Engineering, University of Wisconsin-Madison, WI, 53705 USA
- School of Pharmacy, University of Wisconsin-Madison, WI, 53705 USA
- College of Biomedical Engineering & Bioinstrumentation, Zhejian University, PRC
| |
Collapse
|
39
|
Bunda S, Wang Y, Mitts TF, Liu P, Arab S, Arabkhari M, Hinek A. Aldosterone stimulates elastogenesis in cardiac fibroblasts via mineralocorticoid receptor-independent action involving the consecutive activation of Galpha13, c-Src, the insulin-like growth factor-I receptor, and phosphatidylinositol 3-kinase/Akt. J Biol Chem 2009; 284:16633-16647. [PMID: 19372600 PMCID: PMC2713569 DOI: 10.1074/jbc.m109.008748] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 04/15/2009] [Indexed: 11/06/2022] Open
Abstract
We previously demonstrated that aldosterone, which stimulates collagen production through the mineralocorticoid receptor (MR)-dependent pathway, also induces elastogenesis via a parallel MR-independent mechanism involving insulin-like growth factor-I receptor (IGF-IR) signaling. The present study provides a more detailed explanation of this signaling pathway. Our data demonstrate that small interfering RNA-driven elimination of MR in cardiac fibroblasts does not inhibit aldosterone-induced IGF-IR phosphorylation and subsequent increase in elastin production. These results exclude the involvement of the MR in aldosterone-induced increases in elastin production. Results of further experiments aimed at identifying the upstream signaling component(s) that might be activated by aldosterone also eliminate the putative involvement of pertussis toxin-sensitive Galphai proteins, which have previously been shown to be responsible for some MR-independent effects of aldosterone. Instead, we found that small interfering RNA-dependent elimination of another heterotrimeric G protein, Galpha13, eliminates aldosterone-induced elastogenesis. We further demonstrate that aldosterone first engages Galpha13 and then promotes its transient interaction with c-Src, which constitutes a prerequisite step for aldosterone-dependent activation of the IGF-IR and propagation of consecutive downstream elastogenic signaling involving phosphatidylinositol 3-kinase/Akt. In summary, the data we present reveal new details of an MR-independent cellular signaling pathway through which aldosterone stimulates elastogenesis in human cardiac fibroblasts.
Collapse
Affiliation(s)
- Severa Bunda
- From the Physiology and Experimental Medicine Program, The Hospital for Sick Children, Department of Laboratory Medicine and Pathobiology, Toronto, Ontario M5G 1X8, Canada
| | - Yanting Wang
- From the Physiology and Experimental Medicine Program, The Hospital for Sick Children, Department of Laboratory Medicine and Pathobiology, Toronto, Ontario M5G 1X8, Canada
| | - Thomas F Mitts
- From the Physiology and Experimental Medicine Program, The Hospital for Sick Children, Department of Laboratory Medicine and Pathobiology, Toronto, Ontario M5G 1X8, Canada
| | - Peter Liu
- Heart and Stroke/Richard Lewar Centre for Excellence, University of Toronto, Toronto, Ontario M5G 1X8, Canada; Toronto General Hospital/University Health Network, Toronto, Ontario M5G 1X8, Canada
| | - Sara Arab
- Toronto General Hospital/University Health Network, Toronto, Ontario M5G 1X8, Canada
| | - Majid Arabkhari
- From the Physiology and Experimental Medicine Program, The Hospital for Sick Children, Department of Laboratory Medicine and Pathobiology, Toronto, Ontario M5G 1X8, Canada
| | - Aleksander Hinek
- From the Physiology and Experimental Medicine Program, The Hospital for Sick Children, Department of Laboratory Medicine and Pathobiology, Toronto, Ontario M5G 1X8, Canada; Heart and Stroke/Richard Lewar Centre for Excellence, University of Toronto, Toronto, Ontario M5G 1X8, Canada.
| |
Collapse
|
40
|
An in vitro analysis of mechanical wounding-induced ligand-independent KGFR activation. J Dermatol Sci 2009; 53:182-91. [DOI: 10.1016/j.jdermsci.2008.10.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2008] [Revised: 10/01/2008] [Accepted: 10/14/2008] [Indexed: 11/20/2022]
|
41
|
Deng K, Mock JR, Greenberg S, van Oers NSC, Hansen EJ. Haemophilus ducreyi LspA proteins are tyrosine phosphorylated by macrophage-encoded protein tyrosine kinases. Infect Immun 2008; 76:4692-702. [PMID: 18678665 PMCID: PMC2546853 DOI: 10.1128/iai.00513-08] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 06/19/2008] [Accepted: 07/28/2008] [Indexed: 01/01/2023] Open
Abstract
The LspA proteins (LspA1 and LspA2) of Haemophilus ducreyi are necessary for this pathogen to inhibit the phagocytic activity of macrophage cell lines, an event that can be correlated with a reduction in the level of active Src family protein tyrosine kinases (PTKs) in these eukaryotic cells. During studies investigating this inhibitory mechanism, it was discovered that the LspA proteins themselves were tyrosine phosphorylated after wild-type H. ducreyi cells were incubated with macrophages. LspA proteins in cell-free concentrated H. ducreyi culture supernatant fluid could also be tyrosine phosphorylated by macrophages. This ability to tyrosine phosphorylate the LspA proteins was not limited to immune cell lineages but could be accomplished by both HeLa and COS-7 cells. Kinase inhibitor studies with macrophages demonstrated that the Src family PTKs were required for this tyrosine phosphorylation activity. In silico methods and site-directed mutagenesis were used to identify EPIYG and EPVYA motifs in LspA1 that contained tyrosines that were targets for phosphorylation. A total of four tyrosines could be phosphorylated in LspA1, with LspA2 containing eight predicted tyrosine phosphorylation motifs. Purified LspA1 fusion proteins containing either the EPIYG or EPVYA motifs were shown to be phosphorylated by purified Src PTK in vitro. Macrophage lysates could also tyrosine phosphorylate the LspA proteins and an LspA1 fusion protein via a mechanism that was dependent on the presence of both divalent cations and ATP. Several motifs known to interact with or otherwise affect eukaryotic kinases were identified in the LspA proteins.
Collapse
Affiliation(s)
- Kaiping Deng
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9048, USA
| | | | | | | | | |
Collapse
|
42
|
Yue X, Dreyfus C, Kong TAN, Zhou R. A subset of signal transduction pathways is required for hippocampal growth cone collapse induced by ephrin-A5. Dev Neurobiol 2008; 68:1269-86. [PMID: 18563700 PMCID: PMC2750894 DOI: 10.1002/dneu.20657] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The Eph family tyrosine kinase receptors and their ligands, ephrins, play key roles in a wide variety of physiological and pathological processes including tissue patterning, angiogenesis, bone development, carcinogenesis, axon guidance, and neural plasticity. However, the signaling mechanisms underlying these diverse functions of Eph receptors have not been well understood. In this study, effects of Eph receptor activation on several important signal transduction pathways are examined. In addition, the roles of these pathways in ephrin-A5-induced growth cone collapse were assessed with a combination of biochemical analyses, pharmacological inhibition, and overexpression of dominant-negative and constitutively active mutants. These analyses showed that ephrin-A5 inhibits Erk activity but activates c-Jun N-terminal kinase. However, regulation of these two pathways is not required for ephrin-A5-induced growth cone collapse in hippocampal neurons. Artificial Erk activation by expression of constitutively active Mek1 and B-Raf failed to block ephrin-A5 effects on growth cones, and inhibitors of the Erk pathway also failed to inhibit collapse by ephrin-A5. Inhibition of JNK had no effects on ephrin-A5-induced growth cone collapse either. In addition, inhibitors to PKA and PI3-K showed no effects on ephrin-A5-induced growth cone collapse. However, pharmacological blockade of phosphotyrosine phosphatase activity, the Src family kinases, cGMP-dependent protein kinase, and myosin light chain kinase significantly inhibited ephrin-A5-induced growth cone collapse. These observations indicate that only a subset of signal transduction pathways is required for ephrin-A5-induced growth cone collapse.
Collapse
Affiliation(s)
- Xin Yue
- Department of Chemical Biology, College of Pharmacy, Rutgers University, Piscataway, NJ 08854
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Piscataway, NJ 08854
| | - Cheryl Dreyfus
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Piscataway, NJ 08854
| | - Tony Ah-Ng Kong
- Department of Pharmaceutics, College of Pharmacy, Rutgers University, Piscataway, NJ 08854
| | - Renping Zhou
- Department of Chemical Biology, College of Pharmacy, Rutgers University, Piscataway, NJ 08854
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Piscataway, NJ 08854
| |
Collapse
|
43
|
Knock GA, Snetkov VA, Shaifta Y, Drndarski S, Ward JPT, Aaronson PI. Role of src-family kinases in hypoxic vasoconstriction of rat pulmonary artery. Cardiovasc Res 2008; 80:453-62. [PMID: 18682436 PMCID: PMC2583063 DOI: 10.1093/cvr/cvn209] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Aims We investigated the role of src-family kinases (srcFKs) in hypoxic pulmonary vasoconstriction (HPV) and how this relates to Rho-kinase-mediated Ca2+ sensitization and changes in intracellular Ca2+ concentration ([Ca2+]i). Methods and results Intra-pulmonary arteries (IPAs) were obtained from male Wistar rats. HPV was induced in myograph-mounted IPAs. Auto-phosphorylation of srcFKs and phosphorylation of the regulatory subunit of myosin phosphatase (MYPT-1) and myosin light-chain (MLC20) in response to hypoxia were determined by western blotting. Translocation of Rho-kinase and effects of siRNA knockdown of src and fyn were examined in cultured pulmonary artery smooth muscle cells (PASMCs). [Ca2+]i was estimated in Fura-PE3-loaded IPA. HPV was inhibited by two blockers of srcFKs, SU6656 and PP2. Hypoxia enhanced phosphorylation of three srcFK proteins at Tyr-416 (60, 59, and 54 kDa, corresponding to src, fyn, and yes, respectively) and enhanced srcFK-dependent tyrosine phosphorylation of multiple target proteins. Hypoxia caused a complex, time-dependent enhancement of MYPT-1 and MLC20 phosphorylation, both in the absence and presence of pre-constriction. The sustained component of this enhancement was blocked by SU6656 and the Rho-kinase inhibitor Y27632. In PASMCs, hypoxia caused translocation of Rho-kinase from the nucleus to the cytoplasm, and this was prevented by anti-src siRNA and to a lesser extent by anti-fyn siRNA. The biphasic increases in [Ca2+]i that accompany HPV were also inhibited by PP2. Conclusion Hypoxia activates srcFKs and triggers protein tyrosine phosphorylation in IPA. Hypoxia-mediated Rho-kinase activation, Ca2+ sensitization, and [Ca2+]i responses are depressed by srcFK inhibitors and/or siRNA knockdown, suggesting a central role of srcFKs in HPV.
Collapse
Affiliation(s)
- Greg A Knock
- Department of Asthma, Allergy and Respiratory Science, School of Medicine, King's College London, Room 3.20, Franklin Wilkins Building, Stamford Street, London SE1 9NH, UK.
| | | | | | | | | | | |
Collapse
|
44
|
Varela M, Golder M, Archer F, de las Heras M, Leroux C, Palmarini M. A large animal model to evaluate the effects of Hsp90 inhibitors for the treatment of lung adenocarcinoma. Virology 2008; 371:206-15. [PMID: 17961623 PMCID: PMC2346560 DOI: 10.1016/j.virol.2007.09.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2007] [Revised: 09/06/2007] [Accepted: 09/24/2007] [Indexed: 10/22/2022]
Abstract
Ovine pulmonary adenocarcinoma (OPA) is a naturally occurring lung cancer of sheep caused by Jaagsiekte sheep retrovirus (JSRV). The JSRV envelope glycoprotein (Env) functions as a dominant oncoprotein in vitro and in vivo. In order to develop the basis for the use of OPA as a lung cancer model, we screened a variety of signal transduction inhibitors for their ability to block transformation by the JSRV Env. Most inhibitors were not effective in blocking JSRV Env-induced transformation. On the contrary, various Hsp90 inhibitors efficiently blocked JSRV transformation. This phenomenon was at least partly due to Akt degradation, which is activated in JSRV-transformed cells. Hsp90 was found expressed in tumor cells of sheep with naturally occurring OPA. In addition, Hsp90 inhibitors specifically inhibited proliferation of immortalized and moreover primary cells derived from OPA tumors. Thus, OPA could be used as a large animal model for comprehensive studies investigating the effects of Hsp90 inhibitors in lung adenocarcinoma.
Collapse
Affiliation(s)
- Mariana Varela
- Institute of Comparative Medicine, University of Glasgow Veterinary School, 464 Bearsden Road, Glasgow, G61 1QH, Scotland, UK
| | | | | | | | | | | |
Collapse
|
45
|
Oishi J, Han X, Kang JH, Asami Y, Mori T, Niidome T, Katayama Y. High-throughput colorimetric detection of tyrosine kinase inhibitors based on the aggregation of gold nanoparticles. Anal Biochem 2008; 373:161-3. [DOI: 10.1016/j.ab.2007.08.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 08/20/2007] [Accepted: 08/27/2007] [Indexed: 10/22/2022]
|
46
|
Kumar A, Wang Y, Lin X, Sun G, Parang K. Synthesis and evaluation of 3-phenylpyrazolo[3,4-d]pyrimidine-peptide conjugates as Src kinase inhibitors. ChemMedChem 2007; 2:1346-1360. [PMID: 17530729 DOI: 10.1002/cmdc.200700074] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Indexed: 11/12/2022]
Abstract
3-Phenylpyrazolo[3,4-d]pyrimidine (PhPP) derivatives substituted with an alkyl or aryl carboxylic acid at the N1-endocyclic amine, such as PhPP-CH(2)COOH (IC(50)=250 microM), and peptides Ac-CIYKYY (IC(50)=400 microM) and Ac-YIYGSFK (IC(50)=570 microM) were weak inhibitors of polyE(4)Y phosphorylation by active c-Src. A series of PhPP-peptide conjugates were synthesized using PhPP as an ATP mimic and CIYKYY or YIYGSFK as a peptide substrate to improve the inhibitory potency against active c-Src kinase. PhPP derivatives were attached to the N terminus or the side chain of amino acids in the peptide template. Two N-terminal substituted conjugates, PhPP-CH(2)CO-CIYKYY (IC(50)=0.38 microM) and PhPP-CH(2)CO-YIYGSFK (IC(50)=2.7 microM), inhibited the polyE(4)Y phosphorylation by active c-Src significantly higher than that of the parent compounds. The conjugation of PhPP with the peptides produced a synergistic inhibition effect possibly through creation of favorable interactions between the conjugate and the kinase domain as shown by molecular modeling studies.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 41 Lower College Road, Kingston, Rhode Island 02881, USA
| | | | | | | | | |
Collapse
|
47
|
Stokowski A, Shi S, Sun T, Bartold PM, Koblar SA, Gronthos S. EphB/ephrin-B interaction mediates adult stem cell attachment, spreading, and migration: implications for dental tissue repair. Stem Cells 2007; 25:156-64. [PMID: 17204606 DOI: 10.1634/stemcells.2006-0373] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human adult dental pulp stem cells (DPSCs) reside predominantly within the perivascular niche of dental pulp and are thought to originate from migrating neural crest cells during development. The Eph family of receptor tyrosine kinases and their ligands, the ephrin molecules, play an essential role in the migration of neural crest cells during development and stem cell niche maintenance. The present study examined the expression and function of the B-subclass Eph/ephrin molecules on DPSCs. Multiple receptors were primarily identified on DPSCs within the perivascular niche, whereas ephrin-B1 and ephrin-B3 were expressed by the surrounding pulp tissue. EphB/ephrin-B bidirectional signaling inhibited cell attachment and spreading, predominately via the mitogen-activated protein kinase (MAPK) pathway for forward signaling and phosphorylation of Src family tyrosine kinases via reverse ephrin-B signaling. DPSC migration was restricted through unidirectional ephrin-B1-activated EphB forward signaling, primarily signaling through the MAPK pathway. Furthermore, we observed that ephrin-B1 was downregulated in diseased adult teeth compared with paired uninjured controls. Collectively, these studies suggest that EphB/ephrin-B molecules play a role in restricting DPSC attachment and migration to maintain DPSCs within their stem cell niche under steady-state conditions. These results may have implications for dental pulp development and regeneration.
Collapse
Affiliation(s)
- Agnieszka Stokowski
- Australian Research Council, Centre for the Molecular Genetics of Development, University of Adelaide, Adelaide, South Australia, Australia
| | | | | | | | | | | |
Collapse
|
48
|
Abourbeh G, Dissoki S, Jacobson O, Litchi A, Ben Daniel R, Laki D, Levitzki A, Mishani E. Evaluation of radiolabeled ML04, a putative irreversible inhibitor of epidermal growth factor receptor, as a bioprobe for PET imaging of EGFR-overexpressing tumors. Nucl Med Biol 2007; 34:55-70. [PMID: 17210462 DOI: 10.1016/j.nucmedbio.2006.10.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Accepted: 10/28/2006] [Indexed: 12/21/2022]
Abstract
Overexpression of epidermal growth factor receptor (EGFR) has been implicated in tumor development and malignancy. Evaluating the degree of EGFR expression in tumors could aid in identifying patients for EGFR-targeted therapies and in monitoring treatment. Nevertheless, no currently available assay can reliably quantify receptor content in tumors. Radiolabeled inhibitors of EGFR-TK could be developed as bioprobes for positron emission tomography imaging. Such imaging agents would not only provide a noninvasive quantitative measurement of EGFR content in tumors but also serve as radionuclide carriers for targeted radiotherapy. The potency, reversibility, selectivity and specific binding characteristics of ML04, an alleged irreversible inhibitor of EGFR, were established in vitro. The distribution of the F-18-labeled compound and the extent of EGFR-specific tumor uptake were evaluated in tumor-bearing mice. ML04 demonstrated potent, irreversible and selective inhibition of EGFR, combined with specific binding to the receptor in intact cells. In vivo distribution of the radiolabeled compound revealed tumor/blood and tumor/muscle activity uptake ratios of about 7 and 5, respectively, 3 h following administration of a radiotracer. Nevertheless, only minor EGFR-specific uptake of the compound was detected in these studies, using either EGFR-negative tumors or blocking studies as controls. To improve the in vivo performance of ML04, administration via prolonged intravenous infusion is proposed. Detailed pharmacokinetic characterization of this bioprobe could assist in the development of a kinetic model that would afford accurate measurement of EGFR content in tumors.
Collapse
Affiliation(s)
- Galith Abourbeh
- Department of Medical Biophysics and Nuclear Medicine, Hadassah Hebrew University, Jerusalem 91120, Israel
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhang J, Kalyankrishna S, Wislez M, Thilaganathan N, Saigal B, Wei W, Ma L, Wistuba II, Johnson FM, Kurie JM. SRC-family kinases are activated in non-small cell lung cancer and promote the survival of epidermal growth factor receptor-dependent cell lines. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:366-76. [PMID: 17200208 PMCID: PMC1762707 DOI: 10.2353/ajpath.2007.060706] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The role of Src-family kinases (SFKs) in non-small cell lung cancer (NSCLC) has not been fully defined. Here we addressed this question by examining SFK phosphorylation in NSCLC biopsy samples and using genetic and pharmacological approaches to inhibit SFK expression and activity in cultured NSCLC cells. Immunohistochemical analysis of NSCLC biopsy samples using a Tyr416 phosphorylation-specific, pan-SFK antibody revealed staining in 123 (33%) of 370 tumors. Because c-Src is known to be both an upstream activator and downstream mediator of epidermal growth factor receptor (EGFR), we next investigated SFK phosphorylation in a panel of NSCLC cell lines, including ones that depend on EGFR for survival. The EGFR-dependent NSCLC cell lines HCC827 and H3255 had increased phosphorylation of SFKs, and treatment of these cells with an SFK inhibitor (PP1 or SKI-606) induced apoptosis. PP1 decreased phosphorylation of EGFR, ErbB2, and ErbB3 and strikingly enhanced apoptosis by gefitinib, an EGFR inhibitor. HCC827 cells transfected with c-Src short hairpin RNA exhibited diminished phosphorylation of EGFR and ErbB2 and decreased sensitivity to apoptosis by PP1 or gefitinib. We conclude that SFKs are activated in NSCLC biopsy samples, promote the survival of EGFR-dependent NSCLC cells, and should be investigated as therapeutic targets in NSCLC patients.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M D Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
The development of tyrosine phosphorylation inhibitors has transformed the approach to cancer therapy and is likely to affect other fields of medicine. In spite of the conservation among protein tyrosine kinases (PTKs), one can develop small molecules that block the activity of a narrow spectrum of PTKs and that exhibit much less toxicity than the currently used chemotherapeutic agents. In this review, we discuss principles for inhibiting specific PTKs. We discuss (a) the birth of the concept of generating targeted, nontoxic signal transduction inhibitors, (b) the potential of substrate-competitive versus the more common ATP-competitive PTK inhibitors, (c) the combination of PTK inhibitors with other signal transduction inhibitors to induce apoptosis-the best way to induce the demise of the cancer cell, and (d) the potential to utilize PTK inhibitors/tyrphostins to attenuate nonmalignant pathological conditions, such as immune disorders, tissue rejection, and restenosis.
Collapse
MESH Headings
- Adenosine Triphosphate/chemistry
- Adenosine Triphosphate/metabolism
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/therapeutic use
- Benzamides
- ErbB Receptors/antagonists & inhibitors
- Erlotinib Hydrochloride
- Fusion Proteins, bcr-abl
- Gefitinib
- Graft Occlusion, Vascular/prevention & control
- Humans
- Imatinib Mesylate
- Janus Kinase 2/antagonists & inhibitors
- Janus Kinase 2/metabolism
- Janus Kinase 3/antagonists & inhibitors
- Janus Kinase 3/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Molecular Structure
- Piperazines/chemistry
- Piperazines/metabolism
- Piperazines/therapeutic use
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/metabolism
- Protein Kinase Inhibitors/therapeutic use
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/metabolism
- Pyrimidines/chemistry
- Pyrimidines/metabolism
- Pyrimidines/therapeutic use
- Quinazolines/chemistry
- Quinazolines/metabolism
- Quinazolines/therapeutic use
- Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors
- Receptors, Platelet-Derived Growth Factor/metabolism
- Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors
- Signal Transduction/physiology
- Tyrphostins/chemistry
- Tyrphostins/metabolism
- Tyrphostins/therapeutic use
Collapse
Affiliation(s)
- Alexander Levitzki
- The Silberman Institute for Life Sciences, Department of Biological Chemistry, The Hebrew University, Givat Ram Campus, Jerusalem 91904, Israel.
| | | |
Collapse
|