scholarly journals SH3-SH2 Domain Orientation in Src Kinases

Structure ◽  
2002 ◽  
Vol 10 (7) ◽  
pp. 901-911 ◽  
Author(s):  
Tobias S Ulmer ◽  
Jörn M Werner ◽  
Iain D Campbell
Biochemistry ◽  
2005 ◽  
Vol 44 (39) ◽  
pp. 13043-13050 ◽  
Author(s):  
Gregor Hofmann ◽  
Kristian Schweimer ◽  
Anke Kiessling ◽  
Edith Hofinger ◽  
Finn Bauer ◽  
...  

2007 ◽  
Vol 14 (1) ◽  
pp. 111-124 ◽  
Author(s):  
Alessia Di Florio ◽  
Gabriele Capurso ◽  
Massimo Milione ◽  
Francesco Panzuto ◽  
Raffaele Geremia ◽  
...  

Pancreatic endocrine tumours (PETs) are rare and ‘indolent’ neoplasms that usually develop metastatic lesions and exhibit poor response to standard medical treatments. Few studies have investigated pathways responsible for PET cell growth and invasion and no alternative therapeutic strategies have been proposed. In a recent microarray analysis for genes up-regulated in PETs, we have described the up-regulation of soluble Src family tyrosine kinases in this neoplasia, which may represent potentially promising candidates for therapy. Herein, we have investigated the expression and function of Src family kinases in PETS and PET cell lines. Western blot analysis indicated that Src is highly abundant in the PET cell lines CM and QGP-1. Immunohistochemistry and Western blot analyses showed that Src is up-regulated also in human PET lesions. Pharmacological inhibition of Src family kinases by the specific inhibitor PP2 strongly interfered with adhesion, spreading and migration of PET cell lines. Accordingly, the actin cytoskeleton was profoundly altered after inhibition of Src kinases, whereas even prolonged incubation with PP2 exerted no effect on cell cycle progression and/or apoptosis of PET cells. A transient increase in tyrosine phosphorylation of a subset of proteins was observed in QGP-1 cells adhering to the plate, with a peak at 75 min after seeding, when approximately 80% of cells were attached. Inhibition of Src kinases caused a dramatic reduction in the phosphorylation of proteins with different molecular weight that were isolated from the cell extracts by anti-phosphotyrosine immunoprecipitation or pull-down with the SH2 domain of Src. Among them, the docking protein p130Cas interacted with Src and is a major substrate of the Src kinases in QGP-1 cells undergoing adhesion. Our results suggest that Src kinases play a specific role during adhesion, spreading and migration of PET cells and may indicate therapeutical approaches directed to limiting the metastatic potential of these cells.


1996 ◽  
Vol 16 (6) ◽  
pp. 2606-2613 ◽  
Author(s):  
K Vuori ◽  
H Hirai ◽  
S Aizawa ◽  
E Ruoslahti

Integrin-mediated cell adhesion triggers intracellular signaling cascades, including tyrosine phosphorylation of intracellular proteins. Among these are the focal adhesion proteins p130cas (Cas) and focal adhesion kinase (FAK). Here we identify the kinase(s) mediating integrin-induced Cas phosphorylation and characterize protein-protein interactions mediated by phosphorylated Cas. We found that expression of a constitutively active FAK in fibroblasts results in a consecutive tyrosine phosphorylation of Cas. This effect required the autophosphorylation site of FAK, which is a binding site for Src family kinases. Integrin-mediated phosphorylation of Cas was not, however, compromised in fibroblasts lacking FAK. In contrast, adhesion-induced tyrosine phosphorylation of Cas was reduced in cells lacking Src, whereas enhanced phosphorylation of Cas was observed Csk- cells, in which Src kinases are activated. These results suggest that Src kinases are responsible for the integrin-mediated tyrosine phosphorylation of Cas. FAK seems not to be necessary for phosphorylation of Cas, but when autophosphorylated, FAK may recruit Src family kinases to phosphorylate Cas. Cas was found to form complexes with Src homology 2 (SH2) domain-containing signaling molecules, such as the SH2/SH3 adapter protein Crk, following integrin-induced tyrosine phosphorylation. Guanine nucleotide exchange factors C3G and Sos were found in the Cas-Crk complex upon integrin ligand binding. These observations suggest that Cas serves as a docking protein and may transduce signals to downstream signaling pathways following integrin-mediated cell adhesion.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1555-1555
Author(s):  
Muneyoshi Futami ◽  
Quan-Sheng Zhu ◽  
Gen-Sheng Feng ◽  
Benjamin Neel ◽  
Seth J. Corey

Abstract Abstract 1555 Hematopoietic cytokine receptors, such as the G-CSFR, use Janus and Src kinases to transduce their signal. Less well known is how the receptors activate these cytosolic protein tyrosine kinases. The phosphorylation/dephosphorylation of inhibitory and stimulatory tyrosine residues of the Src kinases are critical regulatory steps. For Lyn, phosphorylation of Y507 inhibits its activity, whereas phosphorylation of Y396 promotes it. We hypothesized that the tyrosine phosphatase SHP2 is activated by G-CSFR signaling, resulting in dephosphorylation at the negative regulatory site Lyn Y507, and that the adaptor protein Gab2 directs SHP2 effects on phospho-Lyn Y507. To address this hypothesis, we established mouse IL-3-dependent Ba/F3 cells which express the G-CSFR (Ba/F3GR cells). (1) After G-CSF stimulation, phosphorylation status of Lyn (Y507, Y396) was determined by immunoblotting, and protein-protein binding between Gab2-SHP2 and Gab2-Lyn was assessed by the immunoprecipitation and the GST-pull down assay. (2) Ba/F3GR cells were transfected with SHP2, Gab2, or the phosphorylation defective Gab2 mutant (Y614F, Y643F), and phosphorylation status of Lyn was determined. (3) G-CSF-dependent proliferation and colony formation was determined after knockdown of Gab2 or SHP2. After stimulation of Ba/F3GR cells with G-CSF, the inhibitory phosphorylation site Lyn Y507 was dephosphorylated, and the activating site Lyn Y396 was phosphorylated, due to the autophosphorylation. This was observed as a spike at 5 min of the G-CSF treatment and returned gradually to the basal state. When SHP2-deficient cells were treated with G-CSF, phospho-Lyn Y507 was not dephosphorylated, whereas a constitutively active SHP2 mutant E76A dephosphorylated Lyn Y507. In addition, when SHP2 was overexpressed in Ba/F3GR cells, prolonged dephosphorylation of Lyn Y507 was observed. These data suggest that SHP2 is involved in the G-CSF-induced activation of Lyn by the dephosphorylation of Lyn Y507. Co-immunoprecipitation studies revealed that an adapter protein Gab2 binds to Lyn, and this binding is abrogated by the G-CSF treatment in a time course similar to the dephosphorylation of phospho-Lyn Y507. Gab2 has two tyrosine residues (Y614, Y643), which, when phosphorylated, provide binding sites for the SH2-domain of SHP2. Engagement of SHP2 through its SH2-phosphotyrosine interaction leads to a conformational change and activates SHP2. We previously reported that G-CSFR signaling phosphorylates Gab2 (Blood 103: 3305, 2004). As expected, wild-type Gab2 co-immunoprecipitated with SHP2, whereas Gab2 double mutant (Y614F, Y643F) did not. When we transfected Ba/F3GR cells with the Gab2 double mutant, G-CSFR-induced dephosphorylation of Lyn Y507 was abrogated. These findings were confirmed by the knockdown experiments using Gab2 siRNA. To examine the biological consequence, G-CSF-dependent proliferation of Ba/F3GR cells was determined. After the knockdown of SHP2 and Gab2, cell proliferation was inhibited significantly (cell number on day 3 was 21% in Ba/F3GR-SHP2 shRNA, and 43% in Ba/F3GR-Gab2 shRNA, compared to Ba/F3GR-control shRNA). Furthermore, G-CSF-induced CFU-G colony formation was inhibited in bone marrow cells from Gab2-/-mice compared to wildtype mice (Gab2-/- 17.7 ± 0.9 vs. wildtype 25.7 ± 3.8). In summary, we found that treatment with G-CSF results in the dephosphorylation of the negative regulatory site Lyn Y507 and activation of Lyn, and that the binding of Gab2 to SHP2 was required for G-CSF-dependent cell proliferation. These data support a mechanistic model for G-CSF-induced proliferation that requires the activation of the Src kinase Lyn through recruitment of tyrosine phosphatase Shp2 via the adaptor protein Gab2. Disclosures: No relevant conflicts of interest to declare.


2014 ◽  
Vol 35 (1) ◽  
pp. 41-51 ◽  
Author(s):  
Huaijian Guo ◽  
Mario-Ernesto Cruz-Munoz ◽  
Ning Wu ◽  
Michael Robbins ◽  
André Veillette

Signaling lymphocytic activation molecule F7 (SLAMF7) is a receptor present on immune cells, including natural killer (NK) cells. It is also expressed on multiple myeloma (MM) cells. This led to development of an anti-SLAMF7 antibody, elotuzumab, showing efficacy against MM. SLAMF7 mediates activating or inhibitory effects in NK cells, depending on whether cells express or do not express the adaptor EAT-2. Since MM cells lack EAT-2, we elucidated the inhibitory effectors of SLAMF7 in EAT-2-negative NK cells and tested whether these effectors were triggered in MM cells. SLAMF7-mediated inhibition in NK cells lacking EAT-2 was mediated by SH2 domain-containing inositol phosphatase 1 (SHIP-1), which was recruited via tyrosine 261 of SLAMF7. Coupling of SLAMF7 to SHIP-1 required Src kinases, which phosphorylated SLAMF7. Although MM cells lack EAT-2, elotuzumab did not induce inhibitory signals in these cells. This was at least partly due to a lack of CD45, a phosphatase required for Src kinase activation. A defect in SLAMF7 function was also observed in CD45-deficient NK cells. Hence, SLAMF7-triggered inhibition is mediated by a mechanism involving Src kinases, CD45, and SHIP-1 that is defective in MM cells. This defect might explain why elotuzumab eliminates MM cells by an indirect mechanism involving the activation of NK cells.


2008 ◽  
Vol 31 (4) ◽  
pp. 23
Author(s):  
Rachel Vanderlaan ◽  
Rod Hardy ◽  
Golam Kabir ◽  
Peter Back ◽  
A J Pawson

Background: ShcA, a scaffolding protein, generates signalspecificity by docking to activated tyrosine kinases through distinct phosphotyrosine recognition motifs, while mediating signal complexity through formation of diverse downstream phosphotyrosine complexes. Mammalian ShcA encodes 3 isoforms having a modular architecture of a PTB domain and SH2 domain, separated by a CH1 region containing tyrosine phosphorylation sites important in Ras-MAPK activation. Objective and Methods: ShcA has a necessary role in cardiovascular development^1,2. However, the role of ShcA in the adult myocardium is largely unknown, also unclear, is how ShcA uses its signaling modules to mediate downstream signaling. To this end, cre/loxP technology was employed to generate a conditional ShcA allele series. The myocardial specific ShcA KO (ShcA CKO) and myocardial restricted domain mutant KI mice were generated using cre expressed from the mlc2v locus^3 coupled with the ShcA floxed allele and in combination with the individual ShcA domain mutant KI alleles^2. Results: ShcACKO mice develop a dilated cardiomyopathy phenotype by 3 months of life, typified by depressed cardiac function and enlarged chamber dimensions. Isolated cardiomyocytes from ShcA CKO mice have preserved contractility indicating an uncoupling between global heart function and single myocyte contractile mechanics. Force-length experiments suggest that the loss of shcAmediates the uncoupling through deregulation of extracellular matrix interactions. Subsequent, analysis of the ShcA myocardial restricted domain mutant KImice suggests that ShcA requires PTB domain docking to upstream tyrosine kinases and subsequent phosphorylation of the CH1 tyrosines important for downstream signaling. Conclusion: ShcA is required for proper maintenance of cardiac function, possibly regulation of extracellular matrix interactions. References: 1. Lai KV, Pawson AJ. The ShcA phosphotyrosine docking protein sensitizescardiovascular signaling in the mouse embryo. Genes and Dev 2000;14:1132-45. 2. Hardy WR. et al. Combinatorial ShcA docking interactions supportdiversity in tissue morphogenesis. Science2007;317:251-6. 3.Minamisawa, s. et al. A post-transcriptional compensatory pathway inheterozygous ventricular myosin light chain 2-deficient mice results in lack ofgene dosage effect during normal cardiac growth or hypertrophy. J Biol Chem 1999;274:10066-70.


Sign in / Sign up

Export Citation Format

Share Document