1
|
Du G, Rao S, Gurbani D, Henning NJ, Jiang J, Che J, Yang A, Ficarro SB, Marto JA, Aguirre AJ, Sorger PK, Westover KD, Zhang T, Gray NS. Structure-Based Design of a Potent and Selective Covalent Inhibitor for SRC Kinase That Targets a P-Loop Cysteine. J Med Chem 2020; 63:1624-1641. [PMID: 31935084 PMCID: PMC7493195 DOI: 10.1021/acs.jmedchem.9b01502] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SRC is a major regulator of many signaling pathways and contributes to cancer development. However, development of a selective SRC inhibitor has been challenging, and FDA-approved SRC inhibitors, dasatinib and bosutinib, are multitargeted kinase inhibitors. Here, we describe our efforts to develop a selective SRC covalent inhibitor by targeting cysteine 277 on the P-loop of SRC. Using a promiscuous covalent kinase inhibitor (CKI) SM1-71 as a starting point, we developed covalent inhibitor 15a, which discriminates SRC from other covalent targets of SM1-71 including TAK1 and FGFR1. As an irreversible covalent inhibitor, compound 15a exhibited sustained inhibition of SRC signaling both in vitro and in vivo. Moreover, 15a exhibited potent antiproliferative effects in nonsmall cell lung cancer cell lines harboring SRC activation, thus providing evidence that this approach may be promising for further drug development efforts.
Collapse
Affiliation(s)
- Guangyan Du
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Suman Rao
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
- Laboratory of Systems Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Deepak Gurbani
- Departments of Biochemistry and Radiation Oncology , The University of Texas Southwestern Medical Center at Dallas , Dallas , Texas 75390 , United States
| | - Nathaniel J Henning
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Jie Jiang
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Jianwei Che
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Annan Yang
- Department of Medical Oncology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Scott B Ficarro
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Jarrod A Marto
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Andrew J Aguirre
- Department of Medical Oncology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Peter K Sorger
- Laboratory of Systems Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Kenneth D Westover
- Departments of Biochemistry and Radiation Oncology , The University of Texas Southwestern Medical Center at Dallas , Dallas , Texas 75390 , United States
| | - Tinghu Zhang
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Nathanael S Gray
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| |
Collapse
|
2
|
Wang Q, Li Y, Zhou J, Liu J, Qin J, Xing F, Zhang J, Cheng J. Clinical significance of Sam68 expression in endometrial carcinoma. Tumour Biol 2015; 36:4509-18. [PMID: 25874492 DOI: 10.1007/s13277-015-3095-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/08/2015] [Indexed: 12/19/2022] Open
Abstract
Sam68 (Src-associated in mitosis of 68 kDa) is a substrate for tyrosine kinase c-Src during mitosis. The nuclear protein level has been found to be associated with progression and prognosis in various human malignant tumors. The aim of this study is to investigate the clinical value of Sam68 in endometrial carcinoma (EC). Sam68 expression was confirmed by real-time PCR, Western blot, and immunofluorescent assay in primary normal endometrial epithelial cells, endometrial carcinoma cell lines, as well as seven pairs of EC and matched adjacent noncancerous endometrial tissues. Moreover, the protein level of Sam68 was evaluated by immunohistochemistry in a cohort of surgical specimens derived from 131 patients including primary endometrial carcinoma (n = 95), endometrial atypical hyperplasia (precancerous lesions, n = 26), and normal endometria (n = 10). In endometrial cancer cell lines, RNA interfering approach was employed to downregulate Sam68 expression to determine its role in proliferation. Clinicopathological relevance and prognostic associations were examined by statistical analyses. Compared with normal endometrial and endometrial atypical hyperplasia tissues, Sam68 significantly elevated in endometrial cancer samples (P < 0.01), which was negative or low in 37 cases (38.9 %) and high in 58 cases (61.1 %). The high expression of Sam68 was associated with histological grade (P < 0.001), FIGO stage (P = 0.039), and myometrial invasion (P = 0.002). Kaplan-Meier analysis demonstrated that overexpression of Sam68 correlated with shorter overall survival. It is confirmed by univariate and multivariate analysis (P < 0.001 and P = 0.048, respectively). Additionally, we found that Sam68 was highly expressed at both the transcriptional and translational levels in endometrial cancer cell lines (Ishikawa, HEC-1B, AN3CA, KLE, and RL95-2) and siRNA knockdown of Sam68 remarkably inhibited cellular proliferation in in vitro models. Sam68 may be useful prognostic marker for EC, and it plays an important role in promoting the cellular proliferation. Further investigation of Sam68 as a potential therapeutic target for EC patients could be of interest.
Collapse
Affiliation(s)
- Qingying Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Middle Road, Shanghai, 200072, China
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Zhang H, Cong H, Song L, Tien P. The nuclear protein Sam68 is redistributed to the cytoplasm and is involved in PI3K/Akt activation during EV71 infection. Virus Res 2014; 180:1-11. [DOI: 10.1016/j.virusres.2013.11.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 11/09/2013] [Accepted: 11/26/2013] [Indexed: 10/25/2022]
|
4
|
Sánchez-Jiménez F, Sánchez-Margalet V. Role of Sam68 in post-transcriptional gene regulation. Int J Mol Sci 2013; 14:23402-23419. [PMID: 24287914 PMCID: PMC3876053 DOI: 10.3390/ijms141223402] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 11/11/2013] [Accepted: 11/13/2013] [Indexed: 01/10/2023] Open
Abstract
The STAR family of proteins links signaling pathways to various aspects of post-transcriptional regulation and processing of RNAs. Sam68 belongs to this class of heteronuclear ribonucleoprotein particle K (hnRNP K) homology (KH) single domain-containing family of RNA-binding proteins that also contains some domains predicted to bind critical components in signal transduction pathways. In response to phosphorylation and other post-transcriptional modifications, Sam68 has been shown to have the ability to link signal transduction pathways to downstream effects regulating RNA metabolism, including transcription, alternative splicing or RNA transport. In addition to its function as a docking protein in some signaling pathways, this prototypic STAR protein has been identified to have a nuclear localization and to take part in the formation of both nuclear and cytosolic multi-molecular complexes such as Sam68 nuclear bodies and stress granules. Coupling with other proteins and RNA targets, Sam68 may play a role in the regulation of differential expression and mRNA processing and translation according to internal and external signals, thus mediating important physiological functions, such as cell death, proliferation or cell differentiation.
Collapse
Affiliation(s)
- Flora Sánchez-Jiménez
- Department of Medical Biochemistry and Molecular Biology and Immunology, UGC Clinical Biochemistry, Virgen Macarena University Hospital, Avenue. Sánchez Pizjuan 4, Medical School, University of Seville, Seville 41009; Spain; E-Mail:
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology and Immunology, UGC Clinical Biochemistry, Virgen Macarena University Hospital, Avenue. Sánchez Pizjuan 4, Medical School, University of Seville, Seville 41009; Spain; E-Mail:
| |
Collapse
|
5
|
Sam68 interacts with IRS1. Biochem Pharmacol 2011; 83:78-87. [PMID: 22005517 DOI: 10.1016/j.bcp.2011.09.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 09/26/2011] [Accepted: 09/28/2011] [Indexed: 12/18/2022]
Abstract
Sam68 (Src associated in mitosis) is a RNA binding protein that links cellular signaling to RNA processing. In previous studies we found that insulin promotes Sam68 relocalization in the cytoplasm allowing Sam68 to associate with p85PI3K, Grb2, GAP and probably the insulin receptor (IR), modulating insulin action positively. In the present work, we wanted to define the role of Sam68 in the first stages of IR signaling. Both BRET and co-immunoprecipitation assays have been used for the study of Sam68 binding to IR, IRS1 and p85-PI3K. BRET saturation experiments indicated, for the first time, that Sam68 associates with IRS1 in basal condition. To map the region of Sam68 implicated in the interaction with IRS1, different Sam68 mutants deleted in the proline-rich domains were used. The deletion of P0, P1 and P2 proline rich domains in N-terminus as well as P4 and P5 in C-terminus of Sam68 increased BRET(50), thus indicating that the affinity of Sam68 for IRS1 is lower when these domains are missing. Moreover, in IR-transfected HEK-293 cells, BRET saturation experiment indicated that insulin increases the affinity between Sam68-Rluc and IRS1-YFP. In conclusion, our data indicate that Sam68 interacts with IRS-1 in basal conditions, and insulin increases the affinity between these two partners.
Collapse
|
6
|
Ishizawar RC, Tice DA, Karaoli T, Parsons SJ. The C terminus of c-Src inhibits breast tumor cell growth by a kinase-independent mechanism. J Biol Chem 2004; 279:23773-81. [PMID: 15031291 DOI: 10.1074/jbc.m312368200] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Overexpression or increased activity of cellular Src (c-Src) is frequently detected in human breast cancer, implicating involvement of c-Src in the etiology of breast carcinomas. Curiously, overexpression of c-Src in tissue culture cells results in a weakly or non-transforming phenotype, indicating that it alone is not sufficient for oncogenesis. However, the protein has been demonstrated to potentiate mitogenic signals from transmembrane receptors. This report investigates the requirement for c-Src in breast cancer as a transducer and integrator of anchorage-dependent and -independent growth signals by utilizing the Src family pharmacological inhibitors, PP1 and PP2, or stable overexpression of the catalytically inactive c-Src mutant (K- c-Src). Both methods of inhibiting endogenous c-Src diminished formation of soft agar colonies and tumors in nude mice. The majority of the dominant-negative activity of K- c-Src was mapped to the Src homology 2 (SH2) domain and C-terminal half of the molecule, but not to the Unique domain, Src homology 3 (SH3) domain, or the N-terminal half of K- c-Src. Further analysis of the C terminus revealed that its ability to inhibit growth localized to the N-terminal lobe (N-lobe) of the catalytic region. These results underscore the requirement for c-Src to maintain the oncogenic phenotype of breast cancer cells and suggest that c-Src may be manipulated to inhibit cell growth by the direct disruption of its catalytic activity or the introduction of either the SH2 domain or the N-lobe of K- c-Src.
Collapse
Affiliation(s)
- Rumey C Ishizawar
- Department of Microbiology and Cancer Center, University of Virginia Health Services, Charlottesville, Virginia 22908, USA
| | | | | | | |
Collapse
|
7
|
Abstract
Sam68 is one of the most studied members of the STAR family of RNA-binding proteins since its identification over a decade ago. Since its ascension into prominence, enormous progress has been made to unmask the link between the RNA-binding properties of Sam68 and the regulation of cellular processes including signal transduction, cell cycle regulation and tumorigenesis and RNA biogenesis in general. In this review we provide a detailed description of the functional domains of Sam68 and the possible biological roles that justify its superSTAR status.
Collapse
Affiliation(s)
- Kiven E Lukong
- Terry Fox Molecular Oncology Group and Bloomfield Center for Research on Aging, Lady Davis Institute for Medical Research, H3T 1E2 Québec, Canada
| | | |
Collapse
|
8
|
Li QH, Haga I, Shimizu T, Itoh M, Kurosaki T, Fujisawa JI. Retardation of the G2-M phase progression on gene disruption of RNA binding protein Sam68 in the DT40 cell line. FEBS Lett 2002; 525:145-50. [PMID: 12163178 DOI: 10.1016/s0014-5793(02)03103-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Sam68 is an RNA binding protein that is tyrosine-phosphorylated by Src during mitosis and has been postulated to have a role in cell cycle control by modulating RNA metabolism. To elucidate the function of this protein, we isolated a Sam68-deficient DT40 cell line by gene disruption. The Sam68-deficient cells exhibited markedly decreased growth and the growth retardation was due to elongation of the G2-M phase, however, the kinase activity associated with Cdc2 remained unaltered. Our results indicate that Sam68 may play a critical role in G2-M progression in a manner independent of the control of Cyclin/Cdc2 kinase activity.
Collapse
Affiliation(s)
- Qing-Hua Li
- Department of Microbiology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi, 570-8506, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
9
|
Morgan MM, Labno CM, Van Seventer GA, Denny MF, Straus DB, Burkhardt JK. Superantigen-induced T cell:B cell conjugation is mediated by LFA-1 and requires signaling through Lck, but not ZAP-70. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5708-18. [PMID: 11698443 DOI: 10.4049/jimmunol.167.10.5708] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The formation of a conjugate between a T cell and an APC requires the activation of integrins on the T cell surface and remodeling of cytoskeletal elements at the cell-cell contact site via inside-out signaling. The early events in this signaling pathway are not well understood, and may differ from the events involved in adhesion to immobilized ligands. We find that conjugate formation between Jurkat T cells and EBV-B cells presenting superantigen is mediated by LFA-1 and absolutely requires Lck. Mutations in the Lck kinase, Src homology 2 or 3 domains, or the myristoylation site all inhibit conjugation to background levels, and adhesion cannot be restored by the expression of Fyn. However, ZAP-70-deficient cells conjugate normally, indicating that Lck is required for LFA-1-dependent adhesion via other downstream pathways. Several drugs that inhibit T cell adhesion to ICAM-1 immobilized on plastic, including inhibitors of mitogen-activated protein/extracellular signal-related kinase kinase, phosphatidylinositol-3 kinase, and calpain, do not inhibit conjugation. Inhibitors of phospholipase C and protein kinase C block conjugation of both wild-type and ZAP-70-deficient cells, suggesting that a phospholipase C that does not depend on ZAP-70 for its activation is involved. These results are not restricted to Jurkat T cells; Ag-specific primary T cell blasts behave similarly. Although the way in which Lck signals to enhance LFA-1-dependent adhesion is not clear, we find that cells lacking functional Lck fail to recruit F-actin and LFA-1 to the T cell:APC contact site, whereas ZAP-70-deficient cells show a milder phenotype characterized by disorganized actin and LFA-1 at the contact site.
Collapse
Affiliation(s)
- M M Morgan
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
10
|
Dankort D, Jeyabalan N, Jones N, Dumont DJ, Muller WJ. Multiple ErbB-2/Neu Phosphorylation Sites Mediate Transformation through Distinct Effector Proteins. J Biol Chem 2001; 276:38921-8. [PMID: 11500516 DOI: 10.1074/jbc.m106239200] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Amplification of the type I receptor tyrosine kinase ErbB-2 (HER2/Neu) is observed in 20-30% of human mammary carcinomas, correlating with a poor clinical prognosis. We have previously demonstrated that four (Tyr(1144), Tyr(1201), Tyr(1226/1227), or Tyr(1253)) of the five known Neu/ErbB-2 autophosphorylation sites can independently mediate transforming signals. The transforming potential of at least two of these autophosphorylation sites (Tyr(1144) and Tyr(1226/1227)) has been further correlated with their ability to associate with Grb2 and Shc adapter proteins, respectively. To confirm the specificity of these interactions, we have created a series of second site mutants in these phosphorylation sites. The results showed that Grb2 recruitment to site 1144 is absolutely required for transforming signal from this autophosphorylation site, whereas association of Shc-mediated transformation is dependent on conservation of the NPXY motif spanning Tyr(1227). A stretch of amino acid identity around tyrosines 1201 (ENPEYLTP)and 1253 (ENPEYLDL) exists, and mutation of key residues within this motif reveals distinct requirements for an intact protein tyrosine-binding protein (NPXY). We show that DOK-R, a protein tyrosine-binding site-containing protein implicated in Ras signaling, interacts with Neu/ErbB-2 at Tyr(1253) as do two unidentified proteins, p150 and p34, the latter correlating with transformation. Together these data argue that ErbB-2/Neu is capable of mediating transformation through distinct effector pathways.
Collapse
Affiliation(s)
- D Dankort
- Institute for Molecular Biology and Biotechnology, Departments of Biology and Pathology, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | | | | | | | | |
Collapse
|
11
|
Liu K, Li L, Nisson PE, Gruber C, Jessee J, Cohen SN. Neoplastic transformation and tumorigenesis associated with sam68 protein deficiency in cultured murine fibroblasts. J Biol Chem 2000; 275:40195-201. [PMID: 11032831 DOI: 10.1074/jbc.m006194200] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Sam68 is a multimeric 68-kDa RNA-binding nuclear protein of unknown function that interacts with, and is tyrosine-phosphorylated by, the oncogenic protein Src during mitosis. Random homozygous knock-out (RHKO) is a retroviral-based antisense RNA strategy that can identify chromosomal genes whose functional disablement leads to reversible tumorigenic capabilities. Here we report that RHKO-induced Sam68 deficiency results in neoplastic transformation of murine NIH3T3 fibroblasts. Whereas simple haploinsufficiency of Sam68 produced by insertion mutagenesis in a single chromosomal allele did not detectably affect cell growth, reduction of Sam68 protein to <25% of the wild type level was associated with anchorage-independent growth, defective contact inhibition, and the ability to form metastatic tumors in nude mice. These properties were reversed by cessation of RHKO inactivation. Our findings, which indicate that the Sam68 protein level can prominently affect cell proliferation, implicate Sam68 function in tumorigenesis. Consistent with these results is evidence that cells undergoing mitosis show a dramatic reduction in the level of Sam68 protein.
Collapse
Affiliation(s)
- K Liu
- Department of Genetics and Department of Medicine, Stanford University School of Medicine, Stanford, California 94305-5120, USA
| | | | | | | | | | | |
Collapse
|
12
|
Abstract
Traditionally, growth factor-coupled signaling to the nucleus has been thought to be primarily directed toward transcriptional regulation. However, there are now increasing indications from a diversity of experimental systems that other aspects of RNA processing, including translation, lifetime and stability, and splicing are under strict growth factor control. In this review, we present the emerging evidence for growth factor signaling pathways that impact on these different RNA processing events. Particularly noteworthy is the realization that growth factor signaling through Ras can effect the regulation of two RNA cap-binding proteins, the cytosolic eIF-4E complex, which is necessary for initiating translation, and the nuclear cap-binding complex, the CBC, which plays a role in cap-dependent pre-mRNA splicing, U snRNA export and 3'-end processing. This, taken together with other findings that demonstrate the ability of stress response pathways and the small G protein, Cdc42, to activate the CBC, raises some interesting possibilities regarding how signaling to the two cellular RNA cap-binding protein complexes may coordinate the growth-coupled regulation of gene expression at the level of RNA processing.
Collapse
Affiliation(s)
- K F Wilson
- Department of Molecular Medicine, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
13
|
Ostrowski J, Schullery DS, Denisenko ON, Higaki Y, Watts J, Aebersold R, Stempka L, Gschwendt M, Bomsztyk K. Role of tyrosine phosphorylation in the regulation of the interaction of heterogenous nuclear ribonucleoprotein K protein with its protein and RNA partners. J Biol Chem 2000; 275:3619-28. [PMID: 10652358 DOI: 10.1074/jbc.275.5.3619] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The heterogeneous nuclear ribonucleoprotein K protein recruits a diversity of molecular partners and may act as a docking platform involved in such processes as transcription, RNA processing, and translation. We show that K protein is tyrosine-phosphorylated in vitro by Src and Lck. Treatment with H(2)O(2)/Na(3)VO(4), which induces oxidative stress, stimulated tyrosine phosphorylation of K protein in cultured cells and in intact livers. Tyrosine phosphorylation increased binding of Lck and the proto-oncoprotein Vav to K protein in vitro. Oxidative stress increased the association of K protein with Lck and Vav, suggesting that tyrosine phosphorylation regulates the ability of K protein to recruit these effectors in vivo. Translation-based assay showed that K protein is constitutively bound to many mRNAs in vivo. Native immunoprecipitated K protein-mRNA complexes were disrupted by tyrosine phosphorylation, suggesting that the in vivo binding of K protein to mRNA may be responsive to the extracellular signals that activate tyrosine kinases. This study shows that tyrosine phosphorylation of K protein regulates K protein-protein and K protein-RNA interactions. These data are consistent with a model in which functional interaction of K protein is responsive to changes in the extracellular environment. Acting as a docking platform, K protein may bridge signal transduction pathways to sites of nucleic acid-dependent process such as transcription, RNA processing, and translation.
Collapse
Affiliation(s)
- J Ostrowski
- Department of Medicine, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
A new subfamily of KH-domain-containing RNA-binding proteins is encoded by genes that are conserved from yeast to humans. Mutations with interesting developmental phenotypes have been identified in Caenorhabditis elegans, Drosophila and mouse. It is hypothesized that these bifunctional proteins provide a rich source of interesting molecular information about development and define a new cellular pathway that links signal transduction directly to RNA metabolism.
Collapse
Affiliation(s)
- C Vernet
- Cell and Molecular Biology Institute, University of Texas at Austin 78712-1064, USA.
| | | |
Collapse
|
15
|
Schmitz U, Ishida M, Berk BC. Angiotensin II stimulates tyrosine phosphorylation of phospholipase C-gamma-associated proteins. Characterization of a c-Src-dependent 97-kD protein in vascular smooth muscle cells. Circ Res 1997; 81:550-7. [PMID: 9314836 DOI: 10.1161/01.res.81.4.550] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Stimulation of phospholipase C-gamma (PLC-gamma) is a critical event in angiotensin II (Ang II) signal transduction. We have previously shown that in rat aortic smooth muscle (RASM) cells Ang II stimulates tyrosine phosphorylation of PLC-gamma via activation of c-Src. Because we failed to demonstrate a direct association between c-Src and PLC-gamma, we hypothesized that a linker protein mediates the interaction between these molecules. To identify PLC-gamma-associated proteins, RASM cells were labeled with [32P]orthophosphate and stimulated with 100 nmol/L Ang II for 5 minutes. PLC-gamma was immunoprecipitated, and associated proteins were characterized by autoradiography and Western blotting with anti-phosphotyrosine antibodies. Ang II stimulated the phosphorylation of 47-, 60-, 84-, and 97-kD PLC-gamma-associated proteins. Because Ang II increased tyrosine phosphorylation of only the 97-kD protein, we characterized p97 further. An important role for Src in tyrosine phosphorylation of p97 was suggested by findings that p97 phosphorylation was inhibited by the selective Src-family kinase inhibitor CP-118,556, diminished in mouse aortic smooth muscle (MASM) cells from c-Src knockout mice compared with wild-type MASM cells, and increased in v-Src-transformed NIH-3T3 cells compared with wild-type NIH-3T3 cells. These studies are the first to define a PLC-gamma-associated protein that may be required for Ang II-mediated signal transduction.
Collapse
Affiliation(s)
- U Schmitz
- Department of Medicine, University of Washington, Seattle 98195-7710, USA
| | | | | |
Collapse
|
16
|
Vuica M, Desiderio S, Schneck JP. Differential effects of B cell receptor and B cell receptor-FcgammaRIIB1 engagement on docking of Csk to GTPase-activating protein (GAP)-associated p62. J Exp Med 1997; 186:259-67. [PMID: 9221755 PMCID: PMC2198989 DOI: 10.1084/jem.186.2.259] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/1997] [Revised: 05/14/1997] [Indexed: 02/04/2023] Open
Abstract
The stimulatory and inhibitory pathways initiated by engagement of stimulatory receptors such as the B cell receptor for antigen (BCR) and inhibitory receptors such as Fcgamma receptors of the IIB1 type (FcgammaRIIB1) intersect in ways that are poorly understood at the molecular level. Because the tyrosine kinase Csk is a potential negative regulator of lymphocyte activation, we examined the effects of BCR and FcgammaRIIB1 engagement on the binding of Csk to phosphotyrosine-containing proteins. Stimulation of a B lymphoma cell line, A20, with intact anti-IgG antibody induced a direct, SH2-mediated association between Csk and a 62-kD phosphotyrosine-containing protein that was identified as RasGTPase-activating protein-associated p62 (GAP-A.p62). In contrast, stimulation of A20 cells with anti-IgG F(ab')2 resulted in little increase in the association of Csk with GAP-A.p62. The effect of FcgammaRIIB1 engagement on this association was abolished by blockade of FcgammaRIIB1 with the monoclonal antibody 2.4G2. Furthermore, the increased association between Csk and GAP-A.p62 seen upon stimulation with intact anti-Ig was abrogated in the FcgammaRIIB1-deficient cell line IIA1.6 and recovered when FcgammaRIIB1 expression was restored by transfection. The differential effects of BCR and BCR-FcgammaRIIB1-mediated signaling on the phosphorylation of GAP-A.p62 and its association with Csk suggest that docking of Csk to GAP-A.p62 may function in the negative regulation of antigen receptor-mediated signals in B cells.
Collapse
Affiliation(s)
- M Vuica
- Department of Pathology and Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| | | | | |
Collapse
|
17
|
Yamanashi Y, Baltimore D. Identification of the Abl- and rasGAP-associated 62 kDa protein as a docking protein, Dok. Cell 1997; 88:205-11. [PMID: 9008161 DOI: 10.1016/s0092-8674(00)81841-3] [Citation(s) in RCA: 290] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A 62 kDa protein is highly phosphorylated in many cells containing activated tyrosine kinases. This protein, characterized mainly by its avid association with rasGAP, has proved elusive. Anti-phosphotyrosine antibody was used to purify p62. From peptide sequence, molecular cloning revealed a cDNA encoding a novel protein, p62dok, with little homology to others but with a prominent set of tyrosines and nearby sequences suggestive of SH2 binding sites. In cells, v-Abl tyrosine kinase binds and strongly phosphorylates p62dok, which then binds rasGAP. A monoclonal antibody, 2C4, to the rasGAP-associated p62 reacts with p62dok. Thus, p62dok appears to be the long-sought major substrate of many tyrosine kinases.
Collapse
Affiliation(s)
- Y Yamanashi
- Department of Biology, Massachusetts Institute of Technology, Cambridge 02139, USA
| | | |
Collapse
|
18
|
Beslu N, LaRose J, Casteran N, Birnbaum D, Lecocq E, Dubreuil P, Rottapel R. Phosphatidylinositol-3' kinase is not required for mitogenesis or internalization of the Flt3/Flk2 receptor tyrosine kinase. J Biol Chem 1996; 271:20075-81. [PMID: 8702727 DOI: 10.1074/jbc.271.33.20075] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Flt3/Flk2 is a receptor tyrosine kinase that is expressed on early hematopoietic progenitor cells. Flt3/Flk2 belongs to a family of receptors, including Kit and colony-stimulating factor-1R, which support growth and differentiation within the hematopoietic system. The Flt3/Flk2 ligand, in combination with other growth factors, stimulates the proliferation of hematopoietic progenitors of both lymphoid and myeloid lineages in vitro. We report that phosphatidylinositol 3'-kinase (PI3K) binds to a unique site in the carboxy tail of murine Flt3/Flk2. In distinction to Kit and colony-stimulating factor-1R, mutant receptors unable to couple to PI3K and expressed in rodent fibroblasts or in the interleukin 3-dependent cell line Ba/F3 provide a mitogenic signal comparable to wild-type receptors. Flt3/Flk2 receptors that do not bind to PI3K also normally down-regulate, a function ascribed to PI3K in the context of other receptor systems. These data point to the existence of other unidentified pathways that, alone or in combination with PI3K, transduce these cellular responses following the activation of Flt3/Flk2.
Collapse
Affiliation(s)
- N Beslu
- Molecular Hematology Laboratory, Unite 119, Institut National de la Santé et de la Recherche Mèdicale, 27 Bd Lei Roure, 13009 Marseille, France
| | | | | | | | | | | | | |
Collapse
|
19
|
Finan PM, Hall A, Kellie S. Sam68 from an immortalised B-cell line associates with a subset of SH3 domains. FEBS Lett 1996; 389:141-4. [PMID: 8766817 DOI: 10.1016/0014-5793(96)00552-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The binding of proteins from an immortalised B-cell line to a panel of SH3 domains was investigated in vitro. One of the most prominent SH3 domain binding proteins was a 68 kD polypeptide which strongly associated with the SH3 domains of c-src, p85a and p47phox and weakly with the SH3 domain of PLCgamma and n-src with undetectable binding to the other SH3 domains tested. Immunoblotting identified this protein as human Sam68. The ability of proline-rich peptides homologous to the Sam68 primary sequence to inhibit the binding of Sam68 to SH3 domains was investigated. Only one peptide inhibited binding of Sam68 to the p85alpha SH3 domain, whereas several peptides inhibited binding of Sam68 to c-src SH3 domain, suggesting that Sam68 uses different proline-rich motifs to bind to different SH3 domains. A peptide derived from residues 32-44 of Sam68 which fits the class II SH3 domain binding consensus sequence inhibited binding of Sam68 to both p85alpha SH3 domain and c-src SH3 domain, but with differential potency, suggesting a differential affinity of these SH3 domains for this proline-rich motif.
Collapse
Affiliation(s)
- P M Finan
- Yamanouchi Research Institute, Littlemore Hospital, Oxford, UK
| | | | | |
Collapse
|
20
|
Odai H, Sasaki K, Hanazono Y, Ueno H, Tanaka T, Miyagawa K, Mitani K, Yazaki Y, Hirai H. c-Cbl is inducibly tyrosine-phosphorylated by epidermal growth factor stimulation in fibroblasts, and constitutively tyrosine-phosphorylated and associated with v-Src in v-src-transformed fibroblasts. Jpn J Cancer Res 1995; 86:1119-26. [PMID: 8635998 PMCID: PMC5920666 DOI: 10.1111/j.1349-7006.1995.tb03303.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The c-cbl gene was cloned as the cellular homolog of the v-cbl oncogene that is the transforming component of a murine tumorigenic retrovirus, CAS NS-1, though the biological roles of c-Cbl remain to be elucidated. We have previously reported that c-Cbl is implicated in the signal transduction triggered by granulocyte-macrophage colony-stimulating factor or erythropoietin in hematopoietic cells. Here, we observed tyrosine phosphorylation of C-cbl in cells expressing epidermal growth factor receptor depending on EGF stimulation and in v-src transformed cells. Furthermore, c-Cbl was revealed to associate with v-Src in vivo. By means of binding experiments using glutathione S-transferase fusion proteins, we have found that the SH2 and SH3 domains of many proteins bind to c-Cbl. These findings strongly suggest that c-Cbl is implicated in a wide variety of signal transduction pathways, including those of EGF receptor and Src protein, as well as in the signaling pathways of hematopoietic cells.
Collapse
Affiliation(s)
- H Odai
- Third Department of Internal Medicine, Faculty of Medicine, University of Tokyo
| | | | | | | | | | | | | | | | | |
Collapse
|