1
|
Alfonzo-Méndez MA, Sochacki KA, Strub MP, Taraska JW. Dual clathrin and integrin signaling systems regulate growth factor receptor activation. Nat Commun 2022; 13:905. [PMID: 35173166 PMCID: PMC8850434 DOI: 10.1038/s41467-022-28373-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/18/2022] [Indexed: 12/20/2022] Open
Abstract
The crosstalk between growth factor and adhesion receptors is key for cell growth and migration. In pathological settings, these receptors are drivers of cancer. Yet, how growth and adhesion signals are spatially organized and integrated is poorly understood. Here we use quantitative fluorescence and electron microscopy to reveal a mechanism where flat clathrin lattices partition and activate growth factor signals via a coordinated response that involves crosstalk between epidermal growth factor receptor (EGFR) and the adhesion receptor β5-integrin. We show that ligand-activated EGFR, Grb2, Src, and β5-integrin are captured by clathrin coated-structures at the plasma membrane. Clathrin structures dramatically grow in response to EGF into large flat plaques and provide a signaling platform that link EGFR and β5-integrin through Src-mediated phosphorylation. Disrupting this EGFR/Src/β5-integrin axis prevents both clathrin plaque growth and dampens receptor signaling. Our study reveals a reciprocal regulation between clathrin lattices and two different receptor systems to coordinate and enhance signaling. These findings have broad implications for the regulation of growth factor signaling, adhesion, and endocytosis.
Collapse
Affiliation(s)
- Marco A Alfonzo-Méndez
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Bethesda, MD, 20892, USA
| | - Kem A Sochacki
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Bethesda, MD, 20892, USA
| | - Marie-Paule Strub
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Bethesda, MD, 20892, USA
| | - Justin W Taraska
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Bethesda, MD, 20892, USA.
| |
Collapse
|
2
|
Ouyang M, Wan R, Qin Q, Peng Q, Wang P, Wu J, Allen M, Shi Y, Laub S, Deng L, Lu S, Wang Y. Sensitive FRET Biosensor Reveals Fyn Kinase Regulation by Submembrane Localization. ACS Sens 2019; 4:76-86. [PMID: 30588803 DOI: 10.1021/acssensors.8b00896] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fyn kinase plays crucial roles in hematology and T cell signaling; however, there are currently limited tools to visualize the dynamic Fyn activity in live cells. Here we developed and characterized a highly sensitive Fyn biosensor based on fluorescence resonance energy transfer (FRET) to monitor Fyn kinase activity in live cells. Our results show that Fyn kinase activity can be induced in both mouse embryonic fibroblasts (MEFs) and T cells by ligand engagement. Two different motifs were further introduced to target the biosensor at the cellular membrane microdomains in MEFs, revealing that the Fyn-tagged biosensor had 70% greater response to growth factor stimulation than the Lyn-tagged version. This suggests that the plasma membrane microdomains can be categorized into different functional subdomains. Further experiments show that while the membrane accessibility is necessary for Fyn activation, the localization of Fyn outside of its microdomains causes its hyperactivity, indicating that membrane microdomains provide a suppressive microenvironment for Fyn regulation in MEFs. Interestingly, a relatively high Fyn activity can be observed at perinuclear regions, further supporting the notion that the membrane microenvironment has a significant impact on the local molecular functions. Our work hence highlights a novel Fyn FRET biosensor for live cell imaging and its application in revealing an intricate submembrane regulation of Fyn in live MEFs.
Collapse
Affiliation(s)
- Mingxing Ouyang
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu Province 213164, China
| | - Rongxue Wan
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Qin Qin
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
| | - Qin Peng
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
| | - Pengzhi Wang
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
| | - Jenny Wu
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
| | - Molly Allen
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
| | - Yiwen Shi
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
| | - Shannon Laub
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
| | - Linhong Deng
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu Province 213164, China
| | - Shaoying Lu
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
| | - Yingxiao Wang
- Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, United States
| |
Collapse
|
3
|
Anton KA, Kajita M, Narumi R, Fujita Y, Tada M. Src-transformed cells hijack mitosis to extrude from the epithelium. Nat Commun 2018; 9:4695. [PMID: 30410020 PMCID: PMC6224566 DOI: 10.1038/s41467-018-07163-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 10/15/2018] [Indexed: 12/19/2022] Open
Abstract
At the initial stage of carcinogenesis single mutated cells appear within an epithelium. Mammalian in vitro experiments show that potentially cancerous cells undergo live apical extrusion from normal monolayers. However, the mechanism underlying this process in vivo remains poorly understood. Mosaic expression of the oncogene vSrc in a simple epithelium of the early zebrafish embryo results in extrusion of transformed cells. Here we find that during extrusion components of the cytokinetic ring are recruited to adherens junctions of transformed cells, forming a misoriented pseudo-cytokinetic ring. As the ring constricts, it separates the basal from the apical part of the cell releasing both from the epithelium. This process requires cell cycle progression and occurs immediately after vSrc-transformed cell enters mitosis. To achieve extrusion, vSrc coordinates cell cycle progression, junctional integrity, cell survival and apicobasal polarity. Without vSrc, modulating these cellular processes reconstitutes vSrc-like extrusion, confirming their sufficiency for this process. Potentially cancerous cells undergo live apical extrusion from normal monolayers and vSrc expression induces this in zebrafish epithelia. Here, the authors show that vSrc coordinates cytokinetic ring formation, cell cycle progression, junctional integrity, cell survival and apicobasal polarity to induce extrusion of transformed cells.
Collapse
Affiliation(s)
- Katarzyna A Anton
- Department of Cell & Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Mihoko Kajita
- Division of Molecular Oncology, Institute for Genetic Medicine, Hokkaido University Graduate School of Chemical Sciences and Engineering, Sapporo, 060-0815, Japan
| | - Rika Narumi
- Division of Molecular Oncology, Institute for Genetic Medicine, Hokkaido University Graduate School of Chemical Sciences and Engineering, Sapporo, 060-0815, Japan
| | - Yasuyuki Fujita
- Division of Molecular Oncology, Institute for Genetic Medicine, Hokkaido University Graduate School of Chemical Sciences and Engineering, Sapporo, 060-0815, Japan
| | - Masazumi Tada
- Department of Cell & Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
4
|
Advani G, Lim YC, Catimel B, Lio DSS, Ng NLY, Chüeh AC, Tran M, Anasir MI, Verkade H, Zhu HJ, Turk BE, Smithgall TE, Ang CS, Griffin M, Cheng HC. Csk-homologous kinase (Chk) is an efficient inhibitor of Src-family kinases but a poor catalyst of phosphorylation of their C-terminal regulatory tyrosine. Cell Commun Signal 2017; 15:29. [PMID: 28784162 PMCID: PMC5547543 DOI: 10.1186/s12964-017-0186-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/28/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND C-terminal Src kinase (Csk) and Csk-homologous kinase (Chk) are the major endogenous inhibitors of Src-family kinases (SFKs). They employ two mechanisms to inhibit SFKs. First, they phosphorylate the C-terminal tail tyrosine which stabilizes SFKs in a closed inactive conformation by engaging the SH2 domain in cis. Second, they employ a non-catalytic inhibitory mechanism involving direct binding of Csk and Chk to the active forms of SFKs that is independent of phosphorylation of their C-terminal tail. Csk and Chk are co-expressed in many cell types. Contributions of the two mechanisms towards the inhibitory activity of Csk and Chk are not fully clear. Furthermore, the determinants in Csk and Chk governing their inhibition of SFKs by the non-catalytic inhibitory mechanism are yet to be defined. METHODS We determined the contributions of the two mechanisms towards the inhibitory activity of Csk and Chk both in vitro and in transduced colorectal cancer cells. Specifically, we assayed the catalytic activities of Csk and Chk in phosphorylating a specific peptide substrate and a recombinant SFK member Src. We employed surface plasmon resonance spectroscopy to measure the kinetic parameters of binding of Csk, Chk and their mutants to a constitutively active mutant of the SFK member Hck. Finally, we determined the effects of expression of recombinant Chk on anchorage-independent growth and SFK catalytic activity in Chk-deficient colorectal cancer cells. RESULTS Our results revealed Csk as a robust enzyme catalysing phosphorylation of the C-terminal tail tyrosine of SFKs but a weak non-catalytic inhibitor of SFKs. In contrast, Chk is a poor catalyst of SFK tail phosphorylation but binds SFKs with high affinity, enabling it to efficiently inhibit SFKs with the non-catalytic inhibitory mechanism both in vitro and in transduced colorectal cancer cells. Further analyses mapped some of the determinants governing this non-catalytic inhibitory mechanism of Chk to its kinase domain. CONCLUSIONS SFKs are activated by different upstream signals to adopt multiple active conformations in cells. SFKs adopting these conformations can effectively be constrained by the two complementary inhibitory mechanisms of Csk and Chk. Furthermore, the lack of this non-catalytic inhibitory mechanism accounts for SFK overactivation in the Chk-deficient colorectal cancer cells.
Collapse
Affiliation(s)
- Gahana Advani
- Department of Biochemistry & Molecular Biology, University of Melbourne, Parkville, VIC 3010 Australia
- Bio21 Biotechnology and Molecular Science Institute, University of Melbourne, Parkville, VIC 3010 Australia
- Cell Signalling Research Laboratories, School of Biomedical Sciences, University of Melbourne, Parkville, VIC 3010 Australia
| | - Ya Chee Lim
- Department of Biochemistry & Molecular Biology, University of Melbourne, Parkville, VIC 3010 Australia
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Bruno Catimel
- Walter and Eliza Hall Institute for Medical Research and Department of Medical Biology, University of Melbourne, Parkville, VIC 3010 Australia
| | - Daisy Sio Seng Lio
- Department of Biochemistry & Molecular Biology, University of Melbourne, Parkville, VIC 3010 Australia
- Bio21 Biotechnology and Molecular Science Institute, University of Melbourne, Parkville, VIC 3010 Australia
- Cell Signalling Research Laboratories, School of Biomedical Sciences, University of Melbourne, Parkville, VIC 3010 Australia
| | - Nadia L. Y. Ng
- Department of Biochemistry & Molecular Biology, University of Melbourne, Parkville, VIC 3010 Australia
- Bio21 Biotechnology and Molecular Science Institute, University of Melbourne, Parkville, VIC 3010 Australia
- Cell Signalling Research Laboratories, School of Biomedical Sciences, University of Melbourne, Parkville, VIC 3010 Australia
| | - Anderly C. Chüeh
- Walter and Eliza Hall Institute for Medical Research and Department of Medical Biology, University of Melbourne, Parkville, VIC 3010 Australia
| | - Mai Tran
- Department of Biochemistry & Molecular Biology, University of Melbourne, Parkville, VIC 3010 Australia
- Bio21 Biotechnology and Molecular Science Institute, University of Melbourne, Parkville, VIC 3010 Australia
| | - Mohd Ishtiaq Anasir
- Department of Biochemistry & Molecular Biology, University of Melbourne, Parkville, VIC 3010 Australia
- Bio21 Biotechnology and Molecular Science Institute, University of Melbourne, Parkville, VIC 3010 Australia
| | - Heather Verkade
- Department of Biochemistry & Molecular Biology, University of Melbourne, Parkville, VIC 3010 Australia
| | - Hong-Jian Zhu
- Department of Surgery, University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3052 Australia
| | - Benjamin E. Turk
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT USA
| | - Thomas E. Smithgall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Ching-Seng Ang
- Bio21 Biotechnology and Molecular Science Institute, University of Melbourne, Parkville, VIC 3010 Australia
| | - Michael Griffin
- Department of Biochemistry & Molecular Biology, University of Melbourne, Parkville, VIC 3010 Australia
- Bio21 Biotechnology and Molecular Science Institute, University of Melbourne, Parkville, VIC 3010 Australia
| | - Heung-Chin Cheng
- Department of Biochemistry & Molecular Biology, University of Melbourne, Parkville, VIC 3010 Australia
- Bio21 Biotechnology and Molecular Science Institute, University of Melbourne, Parkville, VIC 3010 Australia
- Cell Signalling Research Laboratories, School of Biomedical Sciences, University of Melbourne, Parkville, VIC 3010 Australia
| |
Collapse
|
5
|
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
|
6
|
|
7
|
The Hypothalamic Proline-Rich Polypeptide-1 (Galarmin) and its Analogue d-15 are the Inhibitors of Protein Tyrosine Kinase Activity at Cyclophosphamide-Induced Lymphocytopenia. Neurochem Res 2012; 37:2-4. [DOI: 10.1007/s11064-011-0579-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 08/04/2011] [Accepted: 08/05/2011] [Indexed: 10/17/2022]
|
8
|
Saminathan H, Asaithambi A, Anantharam V, Kanthasamy AG, Kanthasamy A. Environmental neurotoxic pesticide dieldrin activates a non receptor tyrosine kinase to promote PKCδ-mediated dopaminergic apoptosis in a dopaminergic neuronal cell model. Neurotoxicology 2011; 32:567-77. [PMID: 21801747 DOI: 10.1016/j.neuro.2011.06.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 06/17/2011] [Accepted: 06/23/2011] [Indexed: 12/31/2022]
Abstract
Oxidative stress and apoptosis are two key pathophysiological mechanisms underlying dopaminergic degeneration in Parkinson's disease (PD). Recently, we identified that proteolytic activation of protein kinase C-delta (PKCδ), a member of the novel PKC family, contributes to oxidative stress-induced dopaminergic degeneration and that phosphorylation of tyrosine residue 311 (tyr311) on PKCδ is a key event preceding the PKCδ proteolytic activation during oxidative damage. Herein, we report that a non-receptor tyrosine kinase Fyn is significantly expressed in a dopaminergic neuronal N27 cell model. Exposure of N27 cells to the dopaminergic toxicant dieldrin (60 μM) rapidly activated Fyn kinase, PKCδ-tyr311 phosphorylation and proteolytic cleavage. Fyn kinase activation precedes the caspase-3-mediated proteolytic activation of PKCδ. Pre-treatment with p60-tyrosine-specific kinase inhibitor (TSKI) almost completely attenuated dieldrin-induced phosphorylation of PKCδ-tyr311 and its proteolytic activation. Additionally, TSKI almost completely blocked dieldrin-induced apoptotic cell death. To further confirm Fyn's role in the pro-apoptotic function of PKCδ, we adopted the RNAi approach. siRNA-mediated knockdown of Fyn kinase also effectively attenuated dieldrin-induced phosphorylation of PKCδ-tyr311, caspase-3-mediated PKCδ proteolytic cleavage, and DNA fragmentation, suggesting that Fyn kinase regulates the pro-apoptotic function of PKCδ. Collectively, these results demonstrate for the first time that Fyn kinase is a pro-apoptotic kinase that regulates upstream signaling of the PKCδ-mediated apoptotic cell death pathway in neurotoxicity models of pesticide exposure.
Collapse
Affiliation(s)
- Hariharan Saminathan
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA 50011, USA
| | | | | | | | | |
Collapse
|
9
|
Fukunishi S, Tsuda Y, Takeshita A, Fukui H, Miyaji K, Fukuda A, Higuchi K. p59fyn is associated with the development of hepatic steatosis due to chronic ethanol consumption. J Clin Biochem Nutr 2011; 49:20-4. [PMID: 21765602 PMCID: PMC3128361 DOI: 10.3164/jcbn.10-115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 10/20/2010] [Indexed: 01/12/2023] Open
Abstract
p59fyn, a protein tyrosine kinase belonging to the src-family, is involved in the regulatory mechanism of acute response to ethanol in the central nervous system. A previous report showed an association between src-family kinase activity and fatty acid oxidation, and it also reported that hepatic free fatty acid levels were low in Fyn-/- mice. We examined, using Fyn-/- mice whether Fyn is also involved in fatty acid metabolism and the development of pathological changes in the liver in response to chronic ethanol consumption. C57BL/6J Fyn-/- and Fyn+/+ mice were fed for 8 weeks with either a liquid diet comprising ethanol or one in which the calories from ethanol were replaced with carbohydrates. Chronic ethanol consumption for 8 weeks resulted in remarkable hepatic steatosis in Fyn+/+ mice but not in Fyn-/- mice. Chronic ethanol consumption induced a significant decrease in hepatic FFA and triglyceride levels in Fyn-/- mice. Levels of interleukin-6, which is associated with the enhancement of fatty acid oxidation, was also increased significantly in the livers of ethanol-fed Fyn-/- mice. The results suggest that Fyn is involved in the enhancement of fatty acid oxidation and the development of hepatic steatosis caused by chronic ethanol consumption.
Collapse
Affiliation(s)
- Shinya Fukunishi
- Department of Gastroenteroloy and Hepatology, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | | | | | | | | | | | | |
Collapse
|
10
|
Perez DI, Palomo V, Pérez C, Gil C, Dans PD, Luque FJ, Conde S, Martínez A. Switching reversibility to irreversibility in glycogen synthase kinase 3 inhibitors: clues for specific design of new compounds. J Med Chem 2011; 54:4042-56. [PMID: 21500862 DOI: 10.1021/jm1016279] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Development of kinase-targeted therapies for central nervous system (CNS) diseases is a great challenge. Glycogen synthase kinase 3 (GSK-3) offers a great potential for severe CNS unmet diseases, being one of the inhibitors on clinical trials for different tauopathies. Following our hypothesis based on the enhanced reactivity of residue Cys199 in the binding site of GSK-3, we examine here the suitability of phenylhalomethylketones as irreversible inhibitors. Our data confirm that the halomethylketone unit is essential for the inhibitory activity. Moreover, addition of the halomethylketone moiety to reversible inhibitors turned them into irreversible inhibitors with IC(50) values in the nanomolar range. Overall, the results point out that these compounds might be useful pharmacological tools to explore physiological and pathological processes related to signaling pathways regulated by GSK-3 opening new avenues for the discovery of novel GSK-3 inhibitors.
Collapse
Affiliation(s)
- Daniel I Perez
- Instituto de Química Medica-CSIC, Juan de la Cierva 3, 28006 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Krapf D, Arcelay E, Wertheimer EV, Sanjay A, Pilder SH, Salicioni AM, Visconti PE. Inhibition of Ser/Thr phosphatases induces capacitation-associated signaling in the presence of Src kinase inhibitors. J Biol Chem 2010; 285:7977-85. [PMID: 20068039 PMCID: PMC2832948 DOI: 10.1074/jbc.m109.085845] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 01/11/2010] [Indexed: 11/06/2022] Open
Abstract
Signaling events leading to mammalian sperm capacitation rely on activation/deactivation of proteins by phosphorylation. This cascade includes soluble adenylyl cyclase, an atypical bicarbonate-stimulated adenylyl cyclase, and is mediated by protein kinase A and the subsequent stimulation of protein tyrosine phosphorylation. Recently, it has been proposed that the capacitation-associated increase in tyrosine phosphorylation is governed by Src tyrosine kinase activity. This conclusion was based mostly on the observation that Src is present in sperm and that the Src kinase family inhibitor SU6656 blocked the capacitation-associated increase in tyrosine phosphorylation. Results in the present manuscript confirmed these observations and provided evidence that these inhibitors were also able to inhibit protein kinase A phosphorylation, sperm motility, and in vitro fertilization. However, the block of capacitation-associated parameters was overcome when sperm were incubated in the presence of Ser/Thr phosphatase inhibitors such as okadaic acid and calyculin-A at concentrations reported to affect only PP2A. Altogether, these data indicate that Src is not directly involved in the observed increase in tyrosine phosphorylation. More importantly, this work presents strong evidence that capacitation is regulated by two parallel pathways. One of them requiring activation of protein kinase A and the second one involving inactivation of Ser/Thr phosphatases.
Collapse
Affiliation(s)
- Dario Krapf
- From the Department of Veterinary and Animal Science, University of Massachusetts, Amherst, Massachusetts 01003 and
| | - Enid Arcelay
- From the Department of Veterinary and Animal Science, University of Massachusetts, Amherst, Massachusetts 01003 and
| | - Eva V. Wertheimer
- From the Department of Veterinary and Animal Science, University of Massachusetts, Amherst, Massachusetts 01003 and
| | - Archana Sanjay
- the Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Stephen H. Pilder
- the Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Ana M. Salicioni
- From the Department of Veterinary and Animal Science, University of Massachusetts, Amherst, Massachusetts 01003 and
| | - Pablo E. Visconti
- From the Department of Veterinary and Animal Science, University of Massachusetts, Amherst, Massachusetts 01003 and
| |
Collapse
|
12
|
Thienylhalomethylketones: Irreversible glycogen synthase kinase 3 inhibitors as useful pharmacological tools. Bioorg Med Chem 2009; 17:6914-25. [DOI: 10.1016/j.bmc.2009.08.042] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 07/21/2009] [Accepted: 08/13/2009] [Indexed: 01/11/2023]
|
13
|
Röglin L, Altenbrunn F, Seitz O. DNA and RNA-controlled switching of protein kinase activity. Chembiochem 2009; 10:758-65. [PMID: 19241406 DOI: 10.1002/cbic.200800771] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Protein switches use the binding energy gained upon recognition of ligands to modulate the conformation and binding properties of protein segments. We explored whether the programmable nucleic acid mediated recognition might be used to design or mimic constraints that limit the conformational freedom of peptide segments. The aim was to design nucleic acid-peptide conjugates in which the peptide portion of the conjugate would change the affinity for a protein target upon hybridization. This approach was used to control the affinity of a PNA-phosphopeptide conjugate for the signal transduction protein Src kinase, which binds the cognate phosphopeptides in a linear conformation. Peptide-nucleic acid arms were attached to known peptide binders. The chimeric molecules were studied in three modes: 1) as single strands, 2) constrained by intermolecular hybridization (duplex formation) and 3) constrained by intramolecular hybridization (hairpin formation). Of note, duplexes that were designed to accommodate bulged peptide structures (for example, in hairpins or bulges) had lower binding affinities than duplexes in which the peptide was allowed to adopt a more relaxed conformation. Greater than 90-fold differences in binding affinities were observed. It was, thus, feasible to make use of DNA hybridization to reversibly switch from no to almost complete inhibition of Src-SH2-peptide binding, and vice versa. A series of DNA and PNA-based hybridization experiments revealed the importance of charges and conformational effects. Nucleic acid mediated switching was extended to the use of RNA; this enabled a regulation of the enzymatic activity of the Src kinase. The proof-of-principle results demonstrate for the first time that PNA-peptide chimeras can transduce changes of the concentration of a given RNA molecule to changes of the activity of a signal transduction enzyme.
Collapse
Affiliation(s)
- Lars Röglin
- Department of Biomedical Engineering, Eindhoven University of Technology, De Wielen 8, Eindhoven, The Netherlands
| | | | | |
Collapse
|
14
|
Liu K, Kalesh KA, Ong LB, Yao SQ. An improved mechanism-based cross-linker for multiplexed kinase detection and inhibition in a complex proteome. Chembiochem 2008; 9:1883-8. [PMID: 18624293 DOI: 10.1002/cbic.200800212] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Kai Liu
- Deptartment of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117557, Singapore
| | | | | | | |
Collapse
|
15
|
Dimmock JR, Chamankhah M, Das U, Zello GA, Quail JW, Yang J, Nienaber KH, Sharma RK, Selvakumar P, Balzarini J, De Clercq E, Stables JP. Cytotoxic and Topographical Properties of 6-Arylidene-2-dimethylaminomethylcyclohexanone Hydrochlorides and Related Compounds. J Enzyme Inhib Med Chem 2008; 19:1-10. [PMID: 15202487 DOI: 10.1080/14756360310001624975] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
A number of 2-arylidenecyclohexanones (1a-h) were converted into the corresponding Mannich bases (2a-h) and (3a,f). Evaluation against murine L1210 cells as well as human Molt 4/C8 and CEM T-lymphocytes revealed the marked cytotoxicity of the Mannich bases and also the fact that almost invariably these compounds were more potent than the precursor enones (1a-h). Further evaluation of most of the Mannich bases towards a panel of nearly 60 human tumour cell lines confirmed their utility as potent cytotoxins. In this assay, the compounds showed growth-inhibiting properties greater than the anticancer alkylator melphalan. QSAR studies revealed that in some cell lines compounds possessing small electron-attracting aryl substituents showed the greatest potencies. Molecular modeling and X-ray crystallography demonstrated that various interatomic distances and torsion angles correlated with cytotoxicity. A representative compound (2a) demonstrated weak inhibiting properties towards human N-myristoyltransferase and stimulated a tyrosine protein kinase. A single dose of 100 mg/kg of most of the compounds did not prove to be lethal in mice.
Collapse
Affiliation(s)
- J R Dimmock
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Das U, Selvakumar P, Sharma RK, Haas TA, Dimmock JR. N-acyl-3,5-bis(arylidene)-4-piperidones and related compounds which stimulate fyn kinase. J Enzyme Inhib Med Chem 2007; 22:451-5. [PMID: 17847712 DOI: 10.1080/14756360701192515] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
This study is part of a long term project designed to explore the hypothesis that stimulation of cancer cells followed by treatment with one or more cytotoxic agents may create greater damage to tumours than to the corresponding normal tissues. The aim of the present investigation was to discover various compounds which stimulate a protein tyrosine kinase, namely fyn kinase. The N-acyl-3,5-bis(arylidene)-4-piperidones and related analogues activated this enzyme using concentrations of 25 microM while representative molecules achieved this result at 0.1 microM. Molecular modelling suggested that the compounds interact transiently with the ATP binding site of fyn kinase thereby enhancing the catalytic phosphorylation of proteins. In the future, candidate antineoplastic agents will be designed which incorporate the structural features of these enzyme stimulators with the goal of their being formed in vitro and in vivo prior to the release of cytotoxins.
Collapse
Affiliation(s)
- Umashankar Das
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| | | | | | | | | |
Collapse
|
17
|
Gujral TS, Singh VK, Jia Z, Mulligan LM. Molecular mechanisms of RET receptor-mediated oncogenesis in multiple endocrine neoplasia 2B. Cancer Res 2006; 66:10741-9. [PMID: 17108110 DOI: 10.1158/0008-5472.can-06-3329] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multiple endocrine neoplasia 2B (MEN 2B) is an inherited syndrome of early onset endocrine tumors and developmental anomalies. The disease is caused primarily by a methionine to threonine substitution of residue 918 in the kinase domain of the RET receptor (2B-RET); however, the molecular mechanisms that lead to the disease phenotype are unclear. In this study, we show that the M918T mutation causes a 10-fold increase in ATP binding affinity and leads to a more stable receptor-ATP complex, relative to the wild-type receptor. Further, the M918T mutation alters local protein conformation, correlating with a partial loss of RET kinase autoinhibition. Finally, we show that 2B-RET can dimerize and become autophosphorylated in the absence of ligand stimulation. Our data suggest that multiple distinct but complementary molecular mechanisms underlie the MEN 2B phenotype and provide potential targets for effective therapeutics for this disease.
Collapse
Affiliation(s)
- Taranjit S Gujral
- Departments of Pathology and Biochemistry, Queen's University, Kingston, Ontario, Canada
| | | | | | | |
Collapse
|
18
|
Kalia LV, Pitcher GM, Pelkey KA, Salter MW. PSD-95 is a negative regulator of the tyrosine kinase Src in the NMDA receptor complex. EMBO J 2006; 25:4971-82. [PMID: 16990796 PMCID: PMC1618112 DOI: 10.1038/sj.emboj.7601342] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2006] [Accepted: 08/18/2006] [Indexed: 11/09/2022] Open
Abstract
The tyrosine kinase Src upregulates the activity of the N-methyl-D-aspartate subtype of glutamate receptor (NMDAR) and tyrosine phosphorylation of this receptor is critical for induction of NMDAR-dependent plasticity of synaptic transmission. A binding partner for Src within the NMDAR complex is the protein PSD-95. Here we demonstrate an interaction of PSD-95 with Src that does not require the well-characterized domains of PSD-95. Rather, we show binding to Src through a 12-amino-acid sequence in the N-terminal region of PSD-95, a region not previously known to participate in protein-protein interactions. This region interacts directly with the Src SH2 domain. Contrary to typical SH2 domain binding, the PSD-95-Src SH2 domain interaction is phosphotyrosine-independent. Binding of the Src-interacting region of PSD-95 inhibits Src kinase activity and reduces NMDAR phosphorylation. Intracellularly administering a peptide matching the Src SH2 domain-interacting region of PSD-95 depresses NMDAR currents in cultured neurons and inhibits induction of long-term potentiation in hippocampus. Thus, the PSD-95-Src SH2 domain interaction suppresses Src-mediated NMDAR upregulation, a finding that may be of broad importance for synaptic transmission and plasticity.
Collapse
Affiliation(s)
- Lorraine V Kalia
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Graham M Pitcher
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Kenneth A Pelkey
- Laboratory of Cellular and Synaptic Neurophysiology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Michael W Salter
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Program in Neurosciences and Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada M5G 1X8. Tel.: +1 416 813 6272; Fax: +1 416 813 7921; E-mail:
| |
Collapse
|
19
|
Nishio H, Takase I, Fukunishi S, Takagi T, Tamura A, Miyazaki T, Suzuki K. Evidence for Involvement of p59fyn in Fasting-Induced Thymic Involution. Scand J Immunol 2005; 62:103-7. [PMID: 16101815 DOI: 10.1111/j.1365-3083.2005.01646.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
p59fyn, a member of the src-family protein tyrosine kinase, is expressed abundantly in thymus. We examined the possible involvement of p59fyn in thymic involution induced by a fasting stress in Fyn-/- mice. An acute 48 h fast resulted in severe atrophy of the thymus and a marked decrease of the total thymocyte number with depletion of the CD4+CD8+[double positive (DP)] population in Fyn+/+ (control) mice. A remarkable increase in terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labelling-positive signals was detected in the fasted group of control mice. However, these findings were not observed in Fyn-/- mice. Interestingly, MRL/MPJ-lpr/lpr, a Fas-deficient model animal, also showed no significant decrease of DP cell numbers in the fasted group. p59fyn is known to interact with Fas signalling, and these findings suggest that p59fyn is involved in fasting-induced thymic involution, raising the possibility that Fas/p59fyn-mediated signalling may, at least partially, be associated with the phenomenon.
Collapse
Affiliation(s)
- H Nishio
- Department of Legal Medicine, Osaka Medical College, Takatsuki, Japan.
| | | | | | | | | | | | | |
Collapse
|
20
|
Suprynowicz FA, Disbrow GL, Simic V, Schlegel R. Are transforming properties of the bovine papillomavirus E5 protein shared by E5 from high-risk human papillomavirus type 16? Virology 2005; 332:102-13. [PMID: 15661144 DOI: 10.1016/j.virol.2004.11.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2004] [Revised: 09/10/2004] [Accepted: 11/10/2004] [Indexed: 11/24/2022]
Abstract
The E5 proteins of bovine papillomavirus type 1 (BPV-1) and human papillomavirus type 16 (HPV-16) are small (44-83 amino acids), hydrophobic polypeptides that localize to membranes of the Golgi apparatus and endoplasmic reticulum, respectively. While the oncogenic properties of BPV-1 E5 have been characterized in detail, less is known about HPV-16 E5 due to its low expression in mammalian cells. Using codon-optimized HPV-16 E5 DNA, we have generated stable fibroblast cell lines that express equivalent levels of epitope-tagged BPV-1 and HPV-16 E5 proteins. In contrast to BPV-1 E5, HPV-16 E5 does not activate growth factor receptors, phosphoinositide 3-kinase or c-Src, and fails to induce focus formation, although it does promote anchorage-independent growth in soft agar. These variant activities are apparently unrelated to differences in intracellular localization of the E5 proteins since retargeting HPV-16 E5 to the Golgi apparatus does not induce focus formation.
Collapse
Affiliation(s)
- Frank A Suprynowicz
- Department of Pathology, Georgetown University Medical School, Preclinical Sciences Building, Room GR10C, 3900 Reservoir Road, NW, Box #571432, Washington, DC 20057, USA
| | | | | | | |
Collapse
|
21
|
Yokoyama T, Kamata Y, Ohtsuki K. Casein kinase 2 (CK2)-mediated reduction of the activities of Src family tyrosine kinases in vitro. Biol Pharm Bull 2005; 27:1895-9. [PMID: 15577202 DOI: 10.1248/bpb.27.1895] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The physiological correlation between casein kinase 2 (CK2) and two Src family tyrosine kinases (Src-TKs, Fyn and Src) was mainly investigated in vitro. It was found that (i) Thr-residues of these two Src-TKs were preferentially phosphorylated by CK2 using [gamma-32P]GTP as a phosphate donor; (ii) this phosphorylation was highly stimulated in the presence of poly-Arg; (iii) full phosphorylation of two Src-TKs by CK2 resulted in significant reduction of their TK activities; and (iv) quercetin (a CK2 inhibitor) inhibited the CK2-mediated reduction of their Src-TK activities in vitro. Under the same experimental conditions, similar results were obtained with Yes. These results suggest that CK2 may be a protein kinase responsible for the suppression of at least three Src-TKs (Fyn, Src and Yes) through the specific phosphorylation of their Thr-residues at the cellular level.
Collapse
Affiliation(s)
- Takamasa Yokoyama
- Genetical Biochemistry and Signal Biology, Graduate School of Medical Sciences, Kitasato University
| | | | | |
Collapse
|
22
|
Han B, Bai XH, Lodyga M, Xu J, Yang BB, Keshavjee S, Post M, Liu M. Conversion of mechanical force into biochemical signaling. J Biol Chem 2004; 279:54793-801. [PMID: 15485829 DOI: 10.1074/jbc.m406880200] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Physical forces play important roles in regulating cell proliferation, differentiation, and death by activating intracellular signal transduction pathways. How cells sense mechanical stimulation, however, is largely unknown. Most studies focus on cellular membrane proteins such as ion channels, integrins, and receptors for growth factors as mechanosensory units. Here we show that mechanical stretch-induced c-Src protein tyrosine kinase activation is mediated through the actin filament-associated protein (AFAP). Distributed along the actin filaments, AFAP can directly active c-Src through binding to its Src homology 3 and/or 2 domains. Mutations at these specific binding sites on AFAP blocked mechanical stretch-induced c-Src activation. Therefore, mechanical force can be transmitted along the cytoskeleton, and interaction between cytoskeletal associated proteins and enzymes related to signal transduction may convert physical forces into biochemical reactions. Cytoskeleton deformation-induced protein-protein interaction via specific binding sites may represent a novel intracellular mechanism for cells to sense mechanical stimulation.
Collapse
Affiliation(s)
- Bing Han
- Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Song Y, Kesuma D, Wang J, Deng Y, Duan J, Wang JH, Qi RZ. Specific inhibition of cyclin-dependent kinases and cell proliferation by harmine. Biochem Biophys Res Commun 2004; 317:128-32. [PMID: 15047157 DOI: 10.1016/j.bbrc.2004.03.019] [Citation(s) in RCA: 140] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2004] [Indexed: 11/22/2022]
Abstract
As key regulators of the cell proliferation cycle, cyclin-dependent kinases (CDKs) are attractive targets for the development of anti-tumor drugs. In the present study, harmine was identified from a collection of herbal compounds to be a specific inhibitor of Cdk1/cyclin B, Cdk2/cyclin A, and Cdk5/p25 with IC50 values at low micromoles. It displayed little effect on other serine/threonine and tyrosine kinases tested. The CDK inhibition by harmine is competitive with ATP-Mg2+, suggesting that it binds to the ATP-Mg2+-binding pocket of CDKs. In cytotoxicity assays, harmine exhibited a strong inhibitory effect on the growth and proliferation of carcinoma cells whereas it had no significant effect on quiescent fibroblasts. Further, harmine was found to block DNA replication in the carcinoma cells. Taken together, harmine is a selective inhibitor of CDKs and cell proliferation.
Collapse
Affiliation(s)
- Yongcheng Song
- Institute of Molecular and Cell Biology, 30 Medical Drive, Singapore 117609, Singapore
| | | | | | | | | | | | | |
Collapse
|
24
|
Vojtechová M, Tuhácková Z, Hlavácek J, Velek J, Sovová V. The v-Src and c-Src tyrosine kinases immunoprecipitated from Rous sarcoma virus-transformed cells display different peptide substrate specificities. Arch Biochem Biophys 2004; 421:277-82. [PMID: 14984208 DOI: 10.1016/j.abb.2003.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In the cells transformed by Rous sarcoma virus (RSV), two Src proteins are expressed: the ubiquitous tyrosine kinase c-Src and the v-Src, the product of the transforming gene of the virus. Using three synthetic peptide substrates widely used for testing Src kinase activity, we show that they are phosphorylated with different efficiencies by the v-Src and c-Src tyrosine kinases immunoprecipitated from the tumor cell line H19. The v-Src displays higher efficiency (Vmax/Km ratio) toward all three peptides used, but the Vmax of v-Src is much lower than Vmax of c-Src with two peptides out of three. This difference in substrate specificity, if ignored, may cause misestimation of the amounts of active c-Src and v-Src in RSV-transformed cells. On the other hand, the different peptide substrate specificities may also reflect different protein substrate specificities of the v-Src and c-Src kinases in vivo.
Collapse
Affiliation(s)
- Martina Vojtechová
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, 16637 Prague 6, Czech Republic.
| | | | | | | | | |
Collapse
|
25
|
Chong YP, Mulhern TD, Zhu HJ, Fujita DJ, Bjorge JD, Tantiongco JP, Sotirellis N, Lio DSS, Scholz G, Cheng HC. A novel non-catalytic mechanism employed by the C-terminal Src-homologous kinase to inhibit Src-family kinase activity. J Biol Chem 2004; 279:20752-66. [PMID: 14985335 DOI: 10.1074/jbc.m309865200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although C-terminal Src kinase (CSK)-homologous kinase (CHK) is generally believed to inactivate Src-family tyrosine kinases (SFKs) by phosphorylating their consensus C-terminal regulatory tyrosine (Tyr(T)), exactly how CHK inactivates SFKs is not fully understood. Herein, we report that in addition to phosphorylating Tyr(T), CHK can inhibit SFKs by a novel non-catalytic mechanism. First, CHK directly binds to the SFK members Hck, Lyn, and Src to form stable protein complexes. The complex formation is mediated by a non-catalytic Tyr(T)-independent mechanism because it occurs even in the absence of ATP or when Tyr(T) of Hck is replaced by phenylalanine. Second, the non-catalytic CHK-SFK interaction alone is sufficient to inactivate SFKs by inhibiting the catalytic activity of autophosphorylated SFKs. Third, CHK and Src co-localize to specific plasma membrane microdomains of rat brain cells, suggesting that CHK is in close proximity to Src such that it can effectively inactivate Src in vivo. Fourth, native CHK.Src complex exists in rat brain, and recombinant CHK.Hck complex exists in transfected HEK293T cells, implying that CHK forms stable complexes with SFKs in vivo. Taken together, our findings suggest that CHK inactivates SFKs (i) by phosphorylating their Tyr(T) and (ii) by this novel Tyr(T)-independent mechanism involving direct binding of CHK to SFKs. It has been documented that autophosphorylated SFKs can still be active, in some cases even when their Tyr(T) is phosphorylated. Thus, the ability of the Tyr(T)-independent mechanism to suppress the activity of both non-phosphorylated and autophosphorylated SFKs represents a fail-safe measure employed by CHK to down-regulate SFK signaling under all circumstances.
Collapse
Affiliation(s)
- Yuh-Ping Chong
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Crean JKG, Finlay D, Murphy M, Moss C, Godson C, Martin F, Brady HR. The role of p42/44 MAPK and protein kinase B in connective tissue growth factor induced extracellular matrix protein production, cell migration, and actin cytoskeletal rearrangement in human mesangial cells. J Biol Chem 2002; 277:44187-94. [PMID: 12218048 DOI: 10.1074/jbc.m203715200] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Connective tissue growth factor (CTGF) is a member of an emerging family of immediate-early gene products that coordinate complex biological processes during differentiation and tissue repair. Here we describe the role of CTGF in integrin-mediated adhesive signaling and the production of extracellular matrix components in human mesangial cells. The addition of CTGF to primary mesangial cells induced fibronectin production, cell migration, and cytoskeletal rearrangement. These functional responses were associated with recruitment of Src and phosphorylation of p42/44 MAPK and protein kinase B. The inhibition of CTGF-induced p42/44 MAPK or phosphatidylinositol 3-kinase (PI3K)/protein kinase B pathway activities abrogated the induction of fibronectin expression. In addition, anti-beta(3) integrin antibodies attenuated the activation of both the p42/44 MAPK and protein kinase B and the increase in fibronectin levels. CTGF also induced mesangial cell migration via a beta(3) integrin-dependent mechanism that was similarly sensitive to the inhibition of the p42/44 MAPK and PI3K pathways, and it promoted the adhesion of the mesangial cells to type I collagen via up-regulation of alpha(1) integrin. Transient actin cytoskeletal disassembly was observed following treatment with the ligand over the course of a 24-h period. CTGF induced the loss of focal adhesions from the mesangial cell as evidenced by the loss of punctate vinculin. However, these processes are p42/44 MAPK and PI3K pathway-independent. Our data support the hypothesis that CTGF mediates a number of its biological effects by the induction of signaling processes via beta(3) integrin. However, others such as actin cytoskeleton disassembly are modulated in a beta(3) integrin/MAPK/PI3K-independent manner, indicating that CTGF is a complex pleiotropic factor with the potential to amplify primary pathophysiological responses.
Collapse
Affiliation(s)
- John K G Crean
- Department of Medicine and Therapeutics, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland.
| | | | | | | | | | | | | |
Collapse
|
27
|
Espiritu DJD, Bernardo AA, Robey RB, Arruda JAL. A central role for Pyk2-Src interaction in coupling diverse stimuli to increased epithelial NBC activity. Am J Physiol Renal Physiol 2002; 283:F663-70. [PMID: 12217857 DOI: 10.1152/ajprenal.00338.2001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Regulation of renal Na-HCO cotransporter (NBC1) activity by cholinergic agonists, ANG II, and acute acidosis (CO(2)) requires both Src family kinase (SFK) and classic MAPK pathway activation. The nonreceptor tyrosine kinase proline-rich tyrosine kinase 2 (Pyk2) couples discrete G protein-coupled receptor and growth factor receptor signaling to SFK activation. We examined the role of Pyk2-SFK interaction in coupling these stimuli to increased NBC1 activity in opossum kidney cells. Carbachol increased tyrosine autophosphorylation of endogenous Pyk2 and ectopically expressed wild-type Pyk2 and were abrogated by kinase-dead mutant (Pyk2-KD) overexpression. Pyk2 phosphorylation was calcium/calmodulin dependent, and Pyk2 associated with Src by means of SH2 domain interaction. Pyk2 phosphorylation and Pyk2-Src interaction by carbachol were mimicked by both ANG II and CO(2). To correlate Pyk2 autophosphorylation and Pyk2-Src interaction with NBC activity, cotransporter activity was measured in untransfected cells and in cells overexpressing Pyk2-KD in the presence or absence of carbachol, ANG II, or CO(2). In Pyk2-KD-overexpressing cells, the effect of carbachol, ANG II, and CO(2) was abolished. We conclude that Pyk2 plays a central role in coupling carbachol, ANG II, and CO(2) to increased NBC activity. This coupling is mediated by Pyk2 autophosphorylation and Pyk2-Src interaction.
Collapse
Affiliation(s)
- Doris Joy D Espiritu
- Section of Nephrology, Department of Medicine, University of Illinois at Chicago, 60612, USA
| | | | | | | |
Collapse
|
28
|
Ethanol-Induced Cas Tyrosine Phosphorylation and Fyn Kinase Activation in Rat Brain. Alcohol Clin Exp Res 2002. [DOI: 10.1097/00000374-200208001-00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Nishio H, Suzuki K. Ethanol-Induced Cas Tyrosine Phosphorylation and Fyn Kinase Activation in Rat Brain. Alcohol Clin Exp Res 2002. [DOI: 10.1111/j.1530-0277.2002.tb02700.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
30
|
Robin P, Boulven I, Desmyter C, Harbon S, Leiber D. ET-1 stimulates ERK signaling pathway through sequential activation of PKC and Src in rat myometrial cells. Am J Physiol Cell Physiol 2002; 283:C251-60. [PMID: 12055094 DOI: 10.1152/ajpcell.00601.2001] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In this study, we analyzed in rat myometrial cells the signaling pathways involved in the endothelin (ET)-1-induced extracellular signal-regulated kinase (ERK) activation required for the induction of DNA synthesis. We found that inhibition of protein kinase C (PKC) by Ro-31-8220 abolished ERK activation. Inhibition of phospholipase C (PLC) by U-73122 or of phosphoinositide (PI) 3-kinase by wortmannin partially reduced ERK activation. A similar partial inhibition was observed after treatment with pertussis toxin or PKC downregulation by phorbol ester treatment. The effect of wortmannin was additive with that produced by PKC downregulation but not with that due to pertussis toxin. These results suggest that both diacylglycerol-sensitive PKC, activated by PLC products, and diacylglycerol-insensitive PKC, possibly activated by a G(i)-PI 3-kinase-dependent process, are involved in ET-1-induced ERK activation. These two pathways were found to be activated mainly through the ET(A) receptor subtype. ET-1 and phorbol ester stimulated Src activity in a PKC-dependent manner, both responses being abolished in the presence of Ro-31-8220. Inhibition of Src kinases by PP1 abrogated phorbol ester- and ET-1-induced ERK activation. Finally, ET-1 activated Ras in a PP1- and Ro-31-8220-sensitive manner. Altogether, our results indicate that ET-1 induces ERK activation in rat myometrial cells through the sequential stimulation of PKC, Src, and Ras.
Collapse
Affiliation(s)
- Philippe Robin
- Laboratoire de Signalisation et Régulations Cellulaires, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8619, Université de Paris-Sud, 91405 Orsay cedex, France
| | | | | | | | | |
Collapse
|
31
|
González-Guerrico AM, Cafferata EG, Radrizzani M, Marcucci F, Gruenert D, Pivetta OH, Favaloro RR, Laguens R, Perrone SV, Gallo GC, Santa-Coloma TA. Tyrosine kinase c-Src constitutes a bridge between cystic fibrosis transmembrane regulator channel failure and MUC1 overexpression in cystic fibrosis. J Biol Chem 2002; 277:17239-47. [PMID: 11872746 DOI: 10.1074/jbc.m112456200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cystic fibrosis (CF), a disease caused by mutations in the cystic fibrosis transmembrane regulator (CFTR) chloride channel, is associated in the respiratory system with the accumulation of mucus and impaired lung function. The role of the CFTR channel in the regulation of the intracellular pathways that determine the overexpression of mucin genes is unknown. Using differential display, we have observed the differential expression of several mRNAs that may correspond to putative CFTR-dependent genes. One of these mRNAs was further characterized, and it corresponds to the tyrosine kinase c-Src. Additional results suggest that c-Src is a central element in the pathway connecting the CFTR channel with MUC1 overexpression and that the overexpression of mucins is a primary response to CFTR malfunction in cystic fibrosis, which occurs even in the absence of bacterial infection.
Collapse
|
32
|
Boulven I, Robin P, Desmyter C, Harbon S, Leiber D. Differential involvement of Src family kinases in pervanadate-mediated responses in rat myometrial cells. Cell Signal 2002; 14:341-9. [PMID: 11858941 DOI: 10.1016/s0898-6568(01)00269-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We previously described that pervanadate, a potent tyrosine phosphatase inhibitor, induced contraction of rat myometrium via phospholipase (PL) C-gamma1 activation [Biol Reprod 54 (1996) 1383]. In this study, we found that pervanadate induced tyrosine phosphorylation of the platelet-derived growth factor (PDGF)-beta receptor, interaction of the phosphorylated PDGF receptor with the phosphorylated PLC-gamma1, production of inositol phosphates (InsPs), extracellular signal-regulated kinase (ERK) activation and DNA synthesis. All these responses were insensitive to PDGF receptor kinase inhibition or PDGF receptor down-regulation. We showed that Src family kinases were activated by pervanadate, and that InsPs production and phosphorylation of both PLC-gamma1 and the PDGF receptor were blocked by PP1, an Src inhibitor. In contrast, the stimulation of ERK by pervanadate was totally refractory to PP1. These results demonstrated that the activation of Src by pervanadate is involved in PLC-gamma1/InsPs signalling but does not play a major role in ERK activation.
Collapse
Affiliation(s)
- Isaline Boulven
- Laboratoire de Signalisation et Régulations Cellulaires, Centre National de la Recherche Scientifique (CNRS) UMR 8619, Bâtiment 430, Université de Paris-Sud, 91405 Orsay cedex, France
| | | | | | | | | |
Collapse
|
33
|
Suprynowicz FA, Baege A, Sunitha I, Schlegel R. c-Src activation by the E5 oncoprotein enables transformation independently of PDGF receptor activation. Oncogene 2002; 21:1695-706. [PMID: 11896601 DOI: 10.1038/sj.onc.1205223] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2001] [Revised: 11/03/2001] [Accepted: 12/03/2001] [Indexed: 11/09/2022]
Abstract
The E5 oncoprotein of bovine papillomavirus type 1 is a Golgi-resident, hydrophobic polypeptide that can transform immortalized fibroblasts by activating endogenous platelet-derived growth factor receptor beta (PDGF-R). However, the existence of E5 mutants that dissociate transformation from PDGF-R activation implies that there are additional mechanism(s) by which E5 can transform cells. We now show that both wt E5, and transforming E5 mutants that are defective for PDGF-R activation, constitutively activate endogenous c-Src in NIH3T3 cell lines to levels normally associated with acute growth factor stimulation. The ubiquitous Src family protein tyrosine kinase (PTK) Fyn is not activated by these E5 constructs, nor are focal adhesion kinase and endogenous receptor PTKs for insulin, epidermal growth factor, basic fibroblast growth factor and insulin-like growth factor. We further demonstrate that transforming activity of the L26A E5 mutant, which is highly defective for PDGF-R activation, depends on its ability to activate Src. L26A E5 does not transform SYF cells that are deficient for Src, Fyn and Yes, unless Src expression is reconstituted, and does not transform NIH3T3 cells in which Src PTK activity is maintained at a basal level by means of kinase-defective K295R Src overexpression.
Collapse
Affiliation(s)
- Frank A Suprynowicz
- Department of Pathology, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | |
Collapse
|
34
|
Aouacheria A, Ory S, Schmitt JR, Rigal D, Jurdic P, Gillet G. p60(v-src) and serum control cell shape and apoptosis via distinct pathways in quail neuroretina cells. Oncogene 2002; 21:1171-86. [PMID: 11850837 DOI: 10.1038/sj.onc.1205170] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2001] [Revised: 10/29/2001] [Accepted: 11/07/2001] [Indexed: 11/09/2022]
Abstract
We made use of QNR cells transformed by a thermosensitive (tsNY68) strain of the Rous sarcoma virus (RSV) to compare the effect of p60(v-src) and serum in cultured nerve cells. In this system, both p60(v-src) heat inactivation and serum removal resulted in growth arrest in G1. In both cases, growth arrest was reversible since cell proliferation was rapidly re-induced following respectively p60v-src renaturation or serum re-addition. However, cells did not fully recover their ability to grow in soft agar, suggesting that, in contrast to the cell cycle machinery, the transforming capacities of these cells have been irreversibly altered. We found that p60(v-src) kinase activity prevented detachment from the substratum and cell death following serum removal. Thermal inactivation of p60(v-src) at restrictive temperature (41.5 degrees C), but not serum removal, resulted in dramatic morphological changes, which occurred 4 h after temperature shift up to 41.5 degrees C. Later on, typical features of apoptotic cells could be observed. Cell death was greatly reduced by the caspase-3 inhibitor ZVAD.FMK, but not by the caspase-1 inhibitor Ac-YVAD.CHO. Together, these results suggested that p60(v-src) and serum factors act on distinct pathways, at least in part. In an attempt to identify the signalling pathways involved in the cell response to p60(v-src) down regulation, we found that Erk and Rac were rapidly inactivated following temperature shift up to 41.5 degrees C. Thus, the combined effects of p60(v-src) and serum factors on the cytoskeleton dynamics and the apoptosis machinery are essential for full neoplastic transformation of neuroretina cells.
Collapse
Affiliation(s)
- Abdel Aouacheria
- Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS-Université Claude Bernard 7, passage du Vercors F69367 Lyon cedex 07, France
| | | | | | | | | | | |
Collapse
|
35
|
Rajala RV, Datla RS, Carlsen SA, Anderson DH, Qi Z, Wang JH, Sharma RK. Phosphorylation of human N-myristoyltransferase by N-myristoylated SRC family tyrosine kinase members. Biochem Biophys Res Commun 2001; 288:233-9. [PMID: 11594778 DOI: 10.1006/bbrc.2001.5758] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
N-Myristoyltransferase (NMT) is an essential eukaryotic enzyme that catalyzes the cotranslational and/or posttranslational transfer of myristate to the amino terminal glycine residue of a number of important proteins especially the non-receptor tyrosine kinases whose activity is important for tumorigenesis. Human NMT was found to be phosphorylated by non-receptor tyrosine kinase family members of Lyn, Fyn and Lck and dephosphorylated by the Ca(2+)/calmodulin-dependent protein phosphatase, calcineurin. Deletion of 149 amino acids from the N-terminal end resulted in the absence of phosphorylation suggesting that the phosphorylation sites are located in the N-terminal end of NMT. Furthermore, a site-directed mutagenesis study indicated that substitution of tyrosine 100 with phenylalanine served NMT as a poor substrate for the Lyn kinase. A synthetic peptide corresponding to the amino-terminal region encompassing tyrosine 100 of NMT served as a good substrate for the Lyn and Fyn kinases. Our studies also indicated that NMT was found to interact with Lyn through its N-terminal end in a phosphorylation-dependent manner. This is the first study demonstrating the cross-talk between NMT and their myristoylated protein substrates in signaling pathways.
Collapse
Affiliation(s)
- R V Rajala
- Department of Pathology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | | | | | | | | | | | |
Collapse
|
36
|
Xue Q, Wainright A, Gangakhedkar S, Gibbons I. Multiplexed enzyme assays in capillary electrophoretic single-use microfluidic devices. Electrophoresis 2001; 22:4000-7. [PMID: 11700732 DOI: 10.1002/1522-2683(200110)22:18<4000::aid-elps4000>3.0.co;2-a] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We describe a method of performing multiple enzyme assays in a single reaction vessel. The resolving power of capillary electrophoresis enables several enzyme assays to be analyzed at high speed in microfluidic arrays. Multiplexed measurement can increase throughput significantly without requiring highly dense microfluidic arrays. Enzyme assays in a multiplexed format for selected kinases in this work show essentially identical performance to assays performed individually. This establishes an approach for screening one compound against multiple enzyme targets simultaneously. Another potential application for performing multiplexed enzyme assay is to study protein-protein (especially enzyme-enzyme) interaction by monitoring the enzymatic activity changes.
Collapse
Affiliation(s)
- Q Xue
- ACLARA BioSciences, Inc, Mountain View, CA, USA.
| | | | | | | |
Collapse
|
37
|
Hattori R, Otani H, Uchiyama T, Imamura H, Cui J, Maulik N, Cordis GA, Zhu L, Das DK. Src tyrosine kinase is the trigger but not the mediator of ischemic preconditioning. Am J Physiol Heart Circ Physiol 2001; 281:H1066-74. [PMID: 11514272 DOI: 10.1152/ajpheart.2001.281.3.h1066] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The signal cascade that triggers and mediates ischemic preconditioning (IPC) remains unclear. The present study investigated the role of the Src family of tyrosine kinases in IPC. Isolated and buffer-perfused rat hearts underwent IPC with three cycles of 5-min ischemia and 5-min reperfusion, followed by 30-min ischemia and 120-min reperfusion. The Src tyrosine kinase family-selective inhibitor PP1 was administered between 45 and 30 min before ischemia (early PP1 treatment) or for 15 min before IPC [early PP1-preconditioning (PC) treatment]. PP1 was also administered for 5 min before the sustained ischemia (late PP1 treatment) or after IPC (late PP1-PC treatment). Src kinase was activated after 30 min of ischemia in both the membrane and cytosolic fractions. Src kinase was also activated by IPC but was attenuated after the sustained ischemia. Early and late PP1 treatment inhibited Src activation after the sustained ischemia and reduced infarct size. Early PP1-PC inhibited Src activation after IPC but not after the sustained ischemia and blocked cardioprotection afforded by IPC. Late PP1-PC treatment abrogated IPC-induced activation of Src and protein kinase C (PKC)-epsilon in the membrane but not in the cytosolic fraction. This treatment modality abrogated Src activation after the sustained ischemia and failed to block cardioprotection afforded by IPC. These results suggest that Src kinase activation mediates ischemic injury but triggers IPC in the position either upstream of or parallel to membrane-associated PKC-epsilon.
Collapse
Affiliation(s)
- R Hattori
- Cardiovascular Division, Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut 06030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cam WR, Masaki T, Shiratori Y, Kato N, Ikenoue T, Okamoto M, Igarashi K, Sano T, Omata M. Reduced C-terminal Src kinase activity is correlated inversely with pp60(c-src) activity in colorectal carcinoma. Cancer 2001; 92:61-70. [PMID: 11443610 DOI: 10.1002/1097-0142(20010701)92:1<61::aid-cncr1292>3.0.co;2-d] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Specific kinase activity of the proto-oncogene product pp60(c-src) is reported to be elevated in patients with carcinoma of the colon, and a novel cytoplasmic protein-tyrosine kinase, C-terminal Src kinase (Csk), has been found to inactivate the members of the Src family protein-tyrosine kinase. In this study, Csk activity and pp60(c-src) activity were examined in colorectal tumors as well as in colon carcinoma cell lines. METHODS Colorectal carcinoma tissue and adjacent nonneoplastic tissue from 24 patients, from 8 colon carcinoma cell lines, and from 1 normal colon cell line were used. The levels of pp60(c-src) and Csk in colorectal tissue and cell lines were analyzed by Western and/or Northern blot analysis, and their kinase activity levels were measured by in-gel kinase assay. RESULTS In the samples from 24 patients with colorectal carcinoma, pp60(c-src) kinase activity and protein levels were increased by 7.8 +/- 0.55 and 2.6 +/- 0.13 times the control levels, respectively. Conversely, the Csk protein level and its kinase activity were reduced by 0.53 +/- 0.08 and 0.53 +/- 0.09 times the control levels, respectively. pp60(c-src) kinase activity was correlated inversely with Csk activity (correlation coefficient = -0.71; P < 0.0001). Of the cell lines, pp60(c-src) kinase activity and protein levels, respectively, were 7.4 +/- 1.22 and 1.86 +/- 0.28 times greater than normal control levels. Csk protein level and kinase activity, respectively, were 0.54 +/- 0.13 and 0.52 +/- 0.11 times less normal control levels and were correlated with mRNA amount. CONCLUSIONS Csk mRNA, protein, and its kinase activity were reduced in colorectal carcinoma and were correlated with pp60(c-src) kinase activity level. The reduced activity of Csk may be involved in the transformation of a subset of colorectal carcinoma.
Collapse
Affiliation(s)
- W R Cam
- Department of Gastroenterology, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Robey RB, Ruiz OS, Baniqued J, Mahmud D, Espiritu DJ, Bernardo AA, Arruda JA. SFKs, Ras, and the classic MAPK pathway couple muscarinic receptor activation to increased Na-HCO(3) cotransport activity in renal epithelial cells. Am J Physiol Renal Physiol 2001; 280:F844-50. [PMID: 11292627 DOI: 10.1152/ajprenal.2001.280.5.f844] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cholinergic agents are known to affect the epithelial transport of H2O and electrolytes in the kidney. In proximal tubule cells, cholinergic agonists increase basolateral Na-HCO(3) cotransport activity via M(1) muscarinic receptor activation. The signaling intermediates that couple these G protein-coupled receptors to cotransporter activation, however, are not well defined. We therefore sought to identify distal effectors of muscarinic receptor activation that contribute to increased NBC activity in cultured proximal tubule cells. As demonstrated previously for acute CO2-regulated cotransport activity, we found that inhibitors of Src family kinases (SFKs) or the classic mitogen-activated protein kinase (MAPK) pathway prevented the stimulation of NBC activity by carbachol. The ability of carbachol to activate Src, as well as the proximal (Raf) and distal [extracellular signal-regulated kinases 1 and 2 (ERK1/2)] elements of the classic MAPK module, was compatible with these findings. Cholinergic stimulation of ERK1/2 activity was also completely prevented by overexpression of a dominant negative mutant of Ras (N17-Ras). Taken together, these findings suggest a requirement for the sequential activation of SFKs, Ras, and the classic MAPK pathway [Raf-->MAPK/ERK kinase (MEK)-->ERK]. These findings provide important insights into the molecular mechanisms underlying cholinergic regulation of NBC activity in renal epithelial cells. They also suggest a specific mechanism whereby cholinergic stimulation of the kidney can contribute to pH homeostasis.
Collapse
Affiliation(s)
- R B Robey
- Section of Nephrology, Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Sharma SV, Oneyama C, Yamashita Y, Nakano H, Sugawara K, Hamada M, Kosaka N, Tamaoki T. UCS15A, a non-kinase inhibitor of Src signal transduction. Oncogene 2001; 20:2068-79. [PMID: 11360191 DOI: 10.1038/sj.onc.1204296] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2000] [Revised: 01/24/2001] [Accepted: 01/25/2001] [Indexed: 11/09/2022]
Abstract
Src tyrosine kinase plays key roles in signal transduction following growth factor stimulation and integrin-mediated cell-substrate adhesion. Since src-signal transduction defects are implicated in a multitude of human diseases, we have sought to develop new ways to identify small molecule inhibitors using a yeast-based, activated-src over-expression system. In the present study, we describe the identification of a unique src-signal transduction inhibitor, UCS15A. UCS15A was found to inhibit the src specific tyrosine phosphorylation of numerous proteins in v-src-transformed cells. Two of these phosphoproteins were identified as bona-fide src substrates, cortactin and Sam68. UCS15A differed from conventional src-inhibitors in that it did not inhibit the tyrosine kinase activity of src. In addition, UCS15A appeared to differ from src-destabilizing agents such as herbimycin and radicicol that destabilize src by interfering with Hsp90. Our studies suggest that UCS15A exerted its src-inhibitory effects by a novel mechanism that involved disruption of protein-protein interactions mediated by src. One of the biological consequences of src-inhibition by UCS15A was its ability to inhibit the bone resorption activity of osteoclasts in vitro. These data suggest that UCS15A may inhibit the bone resorption activity of osteoclasts, not by inhibiting src tyrosine kinase activity, but by disrupting the interaction of proteins associated with src, thereby modulating downstream events in the src signal transduction pathway.
Collapse
Affiliation(s)
- S V Sharma
- Tokyo Research Laboratories, Kyowa Hakko Kogyo Co., Ltd. 3-6-6 Asahi-cho, Machida-shi, Tokyo 194, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Affiliation(s)
- S Pestka
- Department of Molecular Genetics and Microbiology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway 08854-5635, USA
| | | | | | | |
Collapse
|
42
|
Nishio H, Matsui K, Tsuji H, Tamura A, Suzuki K. Possible involvement of Fyn kinase in ethanol-stimulated Cas tyrosine phosphorylation in rat cerebellum and cerebral cortex. J Neurochem 2001; 76:1073-9. [PMID: 11181827 DOI: 10.1046/j.1471-4159.2001.00143.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the present study, we have investigated the effect of intraperitoneal injection of ethanol (3.5 g/kg) on tyrosine phosphorylation in rat brain. Immunoblot analysis using an antiphosphotyrosine antibody revealed that a 130-kDa protein band was detected in the brain extract in response to ethanol administration. This ethanol-stimulated tyrosine phosphorylation of the 130-kDa protein was found in the brain but not in the heart, liver or thymus. The 130-kDa phosphotyrosine-containing protein was identified by immunoprecipitation to be Cas, a crk-associated src substrate. This ethanol-stimulated tyrosine phosphorylation of Cas was observed most prominently in the cerebellum and the cerebral cortex. We further examined the possible involvement of Fyn kinase in ethanol-stimulated Cas tyrosine phosphorylation. Immunecomplex kinase assay showed that Fyn was activated in the cerebellum and cerebral cortex of ethanol-administered rats. Immunoprecipitation experiments also showed that Fyn was co-immunoprecipitated with an anti-Cas antibody in these regions from ethanol-administered rats. Furthermore, exogenous Fyn was shown to phosphorylate Cas from cerebellum and cerebral cortex in vitro. These findings indicate that ethanol stimulates tyrosine phosphorylation of Cas in rat cerebellum and cerebral cortex, and that Fyn may be involved in the process.
Collapse
Affiliation(s)
- H Nishio
- Department of Legal Medicine, Osaka Medical College, Takatsuki, Japan
| | | | | | | | | |
Collapse
|
43
|
Nitabach MN, Llamas DA, Araneda RC, Intile JL, Thompson IJ, Zhou YI, Holmes TC. A mechanism for combinatorial regulation of electrical activity: Potassium channel subunits capable of functioning as Src homology 3-dependent adaptors. Proc Natl Acad Sci U S A 2001; 98:705-10. [PMID: 11149959 PMCID: PMC14652 DOI: 10.1073/pnas.98.2.705] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
It is an open question how ion channel subunits that lack protein-protein binding motifs become targeted and covalently modified by cellular signaling enzymes. Here, we show that Src-family protein tyrosine kinases (PTKs) bind to heteromultimeric Shaker-family voltage-gated potassium (Kv) channels by interactions between the Src homology 3 (SH3) domain and the proline-rich SH3 domain ligand sequence in the Shaker-family subunit Kv1.5. Once bound to Kv1.5, Src-family PTKs phosphorylate adjacent subunits in the Kv channel heteromultimer that lack proline-rich SH3 domain ligand sequences. This SH3-dependent tyrosine phosphorylation contributes to significant suppression of voltage-evoked currents flowing through the heteromultimeric channel. These results demonstrate that Kv1.5 subunits function as SH3-dependent adaptor proteins that marshal Src-family kinases to heteromultimeric potassium channel signaling complexes, and thereby confer functional sensitivity upon coassembled channel subunits that are themselves not bound directly to Src-family kinases by allowing their phosphorylation. This is a mechanism for information transfer between subunits in heteromultimeric ion channels that is likely to underlie the generation of combinatorial signaling diversity in the control of cellular electrical excitability.
Collapse
Affiliation(s)
- M N Nitabach
- Department of Biology, New York University, New York, NY 10003, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Bjorge JD, Pang A, Fujita DJ. Identification of protein-tyrosine phosphatase 1B as the major tyrosine phosphatase activity capable of dephosphorylating and activating c-Src in several human breast cancer cell lines. J Biol Chem 2000; 275:41439-46. [PMID: 11007774 DOI: 10.1074/jbc.m004852200] [Citation(s) in RCA: 237] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
c-Src tyrosine kinase activity is elevated in several types of human cancer, and this has been attributed to elevated c-Src expression levels, increased c-Src specific activity, and activating mutations in c-Src. We have found a number of human breast cancer cell lines with elevated c-Src specific activity that also possess elevated phosphatase activity directed against the carboxyl-terminal negative regulatory domain of Src family kinases. To identify this phosphatase, cell extracts from MDA-MB-435S cells were chromatographed and the fractions were assayed for phosphatase activity. Four peaks of phosphatase activity directed against the nonspecific substrate poly(Glu/Tyr) were detected. One peak also dephosphorylated a peptide modeled against the c-Src carboxyl-terminal negative regulatory domain and intact human c-Src. Immunoblotting and immunodepletion experiments identified the phosphatase as protein-tyrosine phosphatase 1B (PTP1B). Examination of several human breast cancer cell lines with increased c-Src activity showed elevated levels of PTP1B protein relative to normal control breast cells. In vitro c-Src reactivation experiments confirmed the ability of PTP1B to dephosphorylate and activate c-Src. In vivo overexpression of PTP1B in 293 cells caused a 2-fold increase of endogenous c-Src kinase activity. Our findings indicate that PTP1B is the primary protein-tyrosine phosphatase capable of dephosphorylating c-Src in several human breast cancer cell lines and suggests a regulatory role for PTP1B in the control of c-Src kinase activity.
Collapse
Affiliation(s)
- J D Bjorge
- Cancer Biology Research Group, Department of Biochemistry and Molecular Biology, University of Calgary Health Sciences Centre, Calgary, Alberta T2N 4N1, Canada
| | | | | |
Collapse
|
45
|
Tan J, Town T, Mullan M. CD45 inhibits CD40L-induced microglial activation via negative regulation of the Src/p44/42 MAPK pathway. J Biol Chem 2000; 275:37224-31. [PMID: 10978311 DOI: 10.1074/jbc.m002006200] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
It has been reported that ligation of CD40 with CD40 ligand (CD40L) results in microglial activation as evidenced by p44/42 mitogen-activated protein kinase (MAPK) dependent tumor necrosis factor alpha (TNF-alpha) production. Previous studies have shown that CD45, a functional transmembrane protein-tyrosine phosphatase, is constitutively expressed at moderate levels on microglial cells and this expression is greatly elevated on activated microglia. To investigate the possibility that CD45 might modulate CD40L-induced microglial activation, we treated primary cultured microglial cells with CD40L and anti-CD45 antibody. Data show that cross-linking of CD45 markedly inhibits CD40L-induced activity of the Src family kinases Lck and Lyn. Further, co-treatment of microglia with CD40L and anti-CD45 antibody results in significant inhibition of microglial TNF-alpha production through inhibition of p44/42 MAPK activity, a downstream signaling event resulting from Src activation. Accordingly, primary cultured microglial cells from mice deficient in CD45 demonstrate hyper-responsiveness to ligation of CD40, as evidenced by increased p44/42 MAPK activation and TNF-alpha production. Taken together, these results show that CD45 plays a novel role in suppressing CD40L-induced microglial activation via negative regulation of the Src/p44/42 MAPK cascade.
Collapse
Affiliation(s)
- J Tan
- Roskamp Institute, Department of Psychiatry, University of South Florida, Tampa, Florida 33613, USA.
| | | | | |
Collapse
|
46
|
Johnson TM, Williamson NA, Scholz G, Jaworowski A, Wettenhall RE, Dunn AR, Cheng HC. Modulation of the catalytic activity of the Src family tyrosine kinase Hck by autophosphorylation at a novel site in the unique domain. J Biol Chem 2000; 275:33353-64. [PMID: 10934191 DOI: 10.1074/jbc.m002022200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Autophosphorylation is a key event in the activation of protein kinases. In this study, we demonstrate that autophosphorylation of the recombinant Src family kinase Hck leads to a 20-fold increase in its specific enzymatic activity. Hck was found to autophosphorylate readily to a stoichiometry of 1.3 mol of phosphate per mol of enzyme, indicating that the kinase autophosphorylated at more than one site. Solid phase sequencing and two-dimensional mapping of the phosphopeptide fragments derived from the autophosphorylated enzyme revealed that the kinase can undergo autophosphorylation at the following two sites: (i) Tyr-388, which is located to the consensus autophosphorylation site commonly found in the activation loop of many protein kinases, and (ii) Tyr-29, which is located in the unique domain of Hck. Hck purified from mouse bone marrow-derived macrophages could also autophosphorylate in vitro at both Tyr-388 and Tyr-29, indicating that naturally occurring Hck can also autophosphorylate at Tyr-29. Furthermore, Hck transiently expressed in human embryonic kidney 293T cells was found to be phosphorylated at Tyr-29 and Tyr-388, proving that Hck can also undergo autophosphorylation at both sites in vivo. The recombinant enzyme carrying the mutation of Tyr-388 to Phe was also able to autophosphorylate at Tyr-29, albeit at a significantly slower rate. A 2-fold increase in the specific enzymatic activity was seen with this mutant despite the stoichiometry of autophosphorylation only approaching 0.2 mol of phosphate per mol of enzyme. This indicates that autophosphorylation of Tyr-29 contributes significantly to the activation of Hck. Regulation of the catalytic activity by phosphorylation of Tyr-29 in the unique domain may represent a new mechanism of regulation of Src family tyrosine kinases.
Collapse
Affiliation(s)
- T M Johnson
- Department of Biochemistry and Molecular Biology, the Department of Medicine, University of Melbourne, Parkville, Victoria 3052, P. O. Australia
| | | | | | | | | | | | | |
Collapse
|
47
|
Ling S, Woronuk G, Sy L, Lev S, Braun AP. Enhanced activity of a large conductance, calcium-sensitive K+ channel in the presence of Src tyrosine kinase. J Biol Chem 2000; 275:30683-9. [PMID: 10893418 DOI: 10.1074/jbc.m004292200] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Large conductance, calcium-sensitive K(+) channels (BK(Ca) channels) contribute to the control of membrane potential in a variety of tissues, including smooth muscle, where they act as the target effector for intracellular "calcium sparks" and the endothelium-derived vasodilator nitric oxide. Various signal transduction pathways, including protein phosphorylation can regulate the activity of BK(Ca) channels, along with many other membrane ion channels. In our study, we have examined the regulation of BK(Ca) channels by the cellular Src gene product (cSrc), a soluble tyrosine kinase that has been implicated in the regulation of both voltage- and ligand-gated ion channels. Using a heterologous expression system, we observed that co-expression of murine BK(Ca) channel and the human cSrc tyrosine kinase in HEK 293 cells led to a calcium-sensitive enhancement of BK(Ca) channel activity in excised membrane patches. In contrast, co-expression with a catalytically inactive cSrc mutant produced no change in BK(Ca) channel activity, demonstrating the requirement for a functional cSrc molecule. Furthermore, we observed that BK(Ca) channels underwent direct tyrosine phosphorylation in cells co-transfected with BK(Ca) channels and active cSrc but not in cells co-transfected with the kinase inactive form of the enzyme. A single Tyr to Phe substitution in the C-terminal half of the channel largely prevented this observed phosphorylation. Given that cSrc may become activated by receptor tyrosine kinases or G-protein-coupled receptors, these findings suggest that cSrc-dependent tyrosine phosphorylation of BK(Ca) channels in situ may represent a novel regulatory mechanism for altering membrane potential and calcium entry.
Collapse
Affiliation(s)
- S Ling
- Smooth Muscle Research Group, Department of Pharmacology and Therapeutics, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | | | | | | | | |
Collapse
|
48
|
Haas M, Askari A, Xie Z. Involvement of Src and epidermal growth factor receptor in the signal-transducing function of Na+/K+-ATPase. J Biol Chem 2000; 275:27832-7. [PMID: 10874030 DOI: 10.1074/jbc.m002951200] [Citation(s) in RCA: 263] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nontoxic concentrations of ouabain, causing partial inhibition of the cardiac myocyte Na(+)/K(+)-ATPase, induce hypertrophy and several growth-related genes through signal pathways that include the activation of Ras and p42/44 mitogen-activated protein kinase (MAPK). The aim of this work was to examine the ouabain-induced events upstream of the Ras/MAPK cascade. Treatment of myocytes with genistein antagonized ouabain-induced activation of the MAPK, suggesting that protein tyrosine phosphorylation has a role. Tyrosine phosphorylation of several myocyte proteins was increased rapidly upon cell exposure to ouabain. Lowering of extracellular K(+) had a similar ouabain-like effect. Ouabain also increased protein tyrosine phosphorylation in A7r5, HeLa, and L929 cells. In cardiac myocytes and A7r5 cells, herbimycin A antagonized the ouabain-induced increase in protein tyrosine phosphorylation and MAPK activation. In both cell types, ouabain stimulated Src kinase activity, Src translocation to the Triton-insoluble fraction, Src association with the epidermal growth factor receptor, and the tyrosine phosphorylation of this receptor on site(s) other than its major autophosphorylation site, Tyr(1173). The findings suggest that (a) the ouabain-induced activation of Src and the Src-induced phosphorylation of the growth factor receptor provide the scaffolding for the recruitment of adaptor proteins and Ras and the activation of Ras/MAPK cascade; and (b) the activation of such pathways may be a common feature of the signal-transducing function of Na(+)/K(+)-ATPase in most cells.
Collapse
Affiliation(s)
- M Haas
- Department of Pharmacology, Medical College of Ohio, Toledo, Ohio 43614, USA
| | | | | |
Collapse
|
49
|
Keshavjee S, Zhang XM, Fischer S, Liu M. Ischemia reperfusion-induced dynamic changes of protein tyrosine phosphorylation during human lung transplantation. Transplantation 2000; 70:525-31. [PMID: 10949198 DOI: 10.1097/00007890-200008150-00022] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We have recently demonstrated that more than 20% of lung cells undergo apoptosis within the first 2 hr of graft reperfusion after human lung transplantation. It has been found that changes of protein tyrosine phosphorylation are involved in the regulation of apoptosis in various cell types. METHODS To determine the protein tyrosine phosphorylation status and related biochemistry changes, lung tissue biopsies were collected from six human lung transplant procedures after cold ischemic preservation (2-5 hr at 4 degrees C), after completing the implantation procedure (approximately 1 hr), and 1 or 2 hr after graft reperfusion. Western blotting was performed to determine protein tyrosine phosphorylation and several signal transduction proteins. Protein tyrosine kinase (PTK) and protein tyrosine phosphatase (PTP) activities were also measured. RESULTS Protein tyrosine phosphorylation was significantly increased after lung implantation and before reperfusion, and significantly decreased during the first 2 hr of graft reperfusion. The activity of Src PTKs was reduced by 50% during graft reperfusion, which was associated with a decrease of Src proteins and human actin filament associated protein, a cofactor for Src activation. PTP activity significantly decreased after lung implantation and remained at a low level 1 hr after reperfusion. After 2 hr of reperfusion, however, PTP activity returned to the basal level. CONCLUSION These dynamic changes of PTK and PTP likely explain the observed alterations of protein tyrosine phosphorylation. The significant decrease in protein tyrosine phosphorylation may be related to the observed apoptotic cell death during human lung transplantation.
Collapse
Affiliation(s)
- S Keshavjee
- Thoracic Surgery Research Laboratory, University Health Network Toronto General Hospital, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
50
|
Sanna PP, Berton F, Cammalleri M, Tallent MK, Siggins GR, Bloom FE, Francesconi W. A role for Src kinase in spontaneous epileptiform activity in the CA3 region of the hippocampus. Proc Natl Acad Sci U S A 2000; 97:8653-7. [PMID: 10890901 PMCID: PMC27003 DOI: 10.1073/pnas.140219097] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Members of the Src family of nonreceptor protein tyrosine kinases (PTKs) have been implicated in the regulation of cellular excitability and synaptic plasticity. We have investigated the role of these PTKs in in vitro models of epileptiform activity. Spontaneous epileptiform discharges were induced in vitro in the CA3 region of rat hippocampal slices by superfusion with the potassium channel blocker 4-aminopyridine in Mg(2+)-free medium. In hippocampal slices treated in this fashion, Src kinase activity was increased and the frequency of epileptiform discharges could be greatly reduced by inhibitor of the Src family of PTKs, 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2), but not by the inactive structural analog 4-amino-7-phenylpyrazol[3,4-d]pyrimidine (PP3). 4-Amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine also reduced epileptiform activity induced by either 4-aminopyridine or Mg(2+)-free medium alone. These observations demonstrate a role for Src family PTKs in the pathophysiology of epilepsy and suggest potential therapeutic targets for antiepileptic therapy.
Collapse
Affiliation(s)
- P P Sanna
- Department of Neuropharmacology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|