The G-CSF Receptor Requires Gab2-Mediated Recruitment of the Tyrosine Phosphatase Shp2 to Promote Lyn-Dependent Proliferation.

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.

2013 ◽  
Vol 200 (4) ◽  
pp. 493-504 ◽  
Author(s):  
Zamal Ahmed ◽  
Chi-Chuan Lin ◽  
Kin M. Suen ◽  
Fernando A. Melo ◽  
James A Levitt ◽  
...  

Constitutive receptor tyrosine kinase phosphorylation requires regulation of kinase and phosphatase activity to prevent aberrant signal transduction. A dynamic mechanism is described here in which the adaptor protein, growth factor receptor–bound protein 2 (Grb2), controls fibroblast growth factor receptor 2 (FGFR2) signaling by regulating receptor kinase and SH2 domain–containing protein tyrosine phosphatase 2 (Shp2) phosphatase activity in the absence of extracellular stimulation. FGFR2 cycles between its kinase-active, partially phosphorylated, nonsignaling state and its Shp2-dephosphorylated state. Concurrently, Shp2 cycles between its FGFR2-phosphorylated and dephosphorylated forms. Both reciprocal activities of FGFR2 and Shp2 were inhibited by binding of Grb2 to the receptor. Phosphorylation of Grb2 by FGFR2 abrogated its binding to the receptor, resulting in up-regulation of both FGFR2’s kinase and Shp2’s phosphatase activity. Dephosphorylation of Grb2 by Shp2 rescued the FGFR2–Grb2 complex. This cycling of enzymatic activity results in a homeostatic, signaling-incompetent state. Growth factor binding perturbs this background cycling, promoting increased FGFR2 phosphorylation and kinase activity, Grb2 dissociation, and downstream signaling. Grb2 therefore exerts constitutive control over the mutually dependent activities of FGFR2 and Shp2.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1163-1163
Author(s):  
Quan-sheng Zhu ◽  
Ling Xia ◽  
Seth Corey

Abstract The Hematopoietin/Cytokine Receptors, such as the Granulocyte Colony-Stimulating Factor (G-CSF) Receptor, signal through the Src and Janus kinases. Src activation involves the sequential regulation of positive and negative tyrosine phosphorylation sites. The C-terminal phosphotyrosine serves as a negative phosphorylation site via its interaction with an endogenous SH2 domain. How Lyn, the predominant Src kinase expressed in granulocytes, becomes activated following G-CSF Receptor engagement is not known. Because Lyn contains a negative phosphotyrosine site (pTyr507), we hypothesize that Shp2, an activating tyrosine phosphatase, might be involved. We report that G-CSF stimulation induces the dissociation of Lyn from the scaffolding protein Gab2, which leads to the dephosphorylation of pTyr507 and autophosphorylation of Tyr396. The dephosphorylation of Lyn pTyr507 was impaired in Shp2-deficient murine embryonic fibroblasts transfected with the G-CSF Receptor in response to G-CSF stimulation. Phosphorylation of Lyn Tyr 396 was impaired in cells treated with Gab2 siRNA. Gab2 constitutively associated with Shp2, and mutations of Tyr614 and Tyr643 in Gab2 abolished the recruitment of Shp2. The constitutively activated Shp2 E76A directed the dephosphorylation of Lyn pTyr507 in vitro. Based on these data, we propose that Gab2 forms a complex with Lyn and following G-CSF stimulation, Gab2 recruits Shp2, which dephosphorylates Lyn pTyr507, leading to Lyn activation. Because gain-of-function mutation in Shp2 or recruitment of Lyn in imatinib-resistant Bcr-Abl+ disease occurs in myeloid and lymphoid leukemias, aberrant pathways involving Src can now be targeted through small molecule inhibitors, such as dasatinib.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3186-3186
Author(s):  
Nicola Esposito ◽  
Irene Colavita ◽  
Fabrizio Quarantelli ◽  
Barbara Izzo ◽  
Luigia Luciano ◽  
...  

Abstract Although rare in chronic phase myeloid leukemia (CML), primary or acquired resistance to the treatment with tyrosine kinase inhibitors (TKI) may be observed in the advanced phases of disease. Bcr/Abl related resistance has been well described, while the other mechanisms of resistance are poorly understood. In this study, we investigated the role of two SH2-containing, non-receptor protein tyrosine phosphatases (Shp1 and Shp2) in the resistance to Imatinib (Ima). To this aim, we have first used, as model system, a couple of Ima-sensitive (KCL22s) and Ima-resistant (KCL22r) KCL22 cell lines. In these cells, Ima resistance is independent by the oncogenic Bcr/Abl activity. We have found a very low level of Shp1 (both mRNA and protein), a protein with a tumour suppressor activity, in the KCL22r resistant cells, when compared to KCL22s sensitive cells. We have al shown the down-regulation of this gene to be related to the methylation level of SHP1 promoter. Indeed, 5-Azacytidine (5-AC) treatment, along with demethylation of the promoter region, re-induced expression of Shp1 in KCL22r. That treatment also re-established the Ima sensitivity, i.e. Ima growth inhibition, in these cells. At molecular level, the restored Ima sensitivity was associated to a significant reduction of phosphorylation of both STAT3 and ERK1/2. To better understand the functional role of Shp1, we carried out mass spectrometry to search for Shp1-binding proteins, and found that Shp1 interacts in these cells with Shp2, a protein phosphatase well known as positive regulator of oncogenic pathways, including the Ras/MAPK pathway. Gain-of-function mutations have been described in various hemopoietic neoplasias including Juvenile Chronic Myelomonocytic Leukemia. In Ph+ cells, oncogenic Bcr/Abl protein activates Shp2 through Gab2, an adaptor protein that, once phosphorylated is able to bind SH2 domain of Shp2. Through complex interactions that may involve the two carboxy-terminal tyrosine residues (542 and 580) Shp2 is also a signal transducer of growth factor receptor. We hypothesized that, Shp1, through dephosphorylation, might modulate the activity of Shp2 and constitute an important mechanism of Ima resistance. Knock-down of Shp1 in KCL22s cell line resulted in complete phosphorylation of Shp2 both 542 and 580 tyrosine residues and in its reduced sensitivity to the drug, thus supporting the role of this protein in Ima sensitivity. On the other hand, knock-down of Shp2 in KCL22r, that shows low Shp1 level, resulted in growth inhibition, restored Ima sensitivity and is associated to a significant reduction of phosphorylation of both STAT3 (60%) and ERK1/2 (70%). The data on primary cells support the role of Shp1 in Ima resistance in patients. Indeed, we analyzed 60 CML patients classified, according to the ENL definitions, as optimal (n =35), suboptimal (n=17) Ima responder, and primary (n=5) or secondary resistant (n=3) to Ima. The levels of Shp1 mRNA were significantly reduced in resistant patients [ratio of SHP1/ABL 3.2 ± 1.04, (mean±SD), *p<0.05] when compared to the suboptimal (3.8±1.54) and optimal responders (5.8±1.77). Moreover, the Shp1 decrease was observed in CD34+ cells isolated from 6 resistant patients in comparison to 6 optimal responders. In conclusion, our study suggests that an aberrant balance between the Shp1 and 2 levels play a role in the Bcr-Abl independent resistance to Ima through activation of Ras/MAPK pathway and that lower levels of Shp1 are associated with non responsive patients.


2000 ◽  
Vol 148 (5) ◽  
pp. 957-970 ◽  
Author(s):  
Valérie Petit ◽  
Brigitte Boyer ◽  
Delphine Lentz ◽  
Christopher E. Turner ◽  
Jean Paul Thiery ◽  
...  

Identification of signaling molecules that regulate cell migration is important for understanding fundamental processes in development and the origin of various pathological conditions. The migration of Nara Bladder Tumor II (NBT-II) cells was used to determine which signaling molecules are specifically involved in the collagen-mediated locomotion. We show here that paxillin is tyrosine phosphorylated after induction of motility on collagen. Overexpression of paxillin mutants in which tyrosine 31 and/or tyrosine 118 were replaced by phenylalanine effectively impaired cell motility. Moreover, stimulation of motility by collagen preferentially enhanced the association of paxillin with the SH2 domain of the adaptor protein CrkII. Mutations in both tyrosine 31 and 118 diminished the phosphotyrosine content of paxillin and prevented the formation of the paxillin–Crk complex, suggesting that this association is necessary for collagen-mediated NBT-II cell migration. Other responses to collagen, such as cell adhesion and spreading, were not affected by these mutations. Overexpression of wild-type paxillin or Crk could bypass the migration-deficient phenotype. Both the SH2 and the SH3 domains of CrkII are shown to play a critical role in this collagen-mediated migration. These results demonstrate the important role of the paxillin–Crk complex in the collagen-induced cell motility.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5106-5106
Author(s):  
Sausan A. Moharram ◽  
Julhash U. Kazi ◽  
Lars Rönnstrand

Abstract The receptor tyrosine kinase FLT3 is found to be a mutated oncogene in hematological malignancies including acute myeloid leukemia (AML). FLT3 inhibitors in combination with chemotherapy display promising results in a clinical setting, but patients relapse after short-term treatment due to the development of resistant disease. Therefore, targeting signaling proteins downstream of FLT3 can be an alternative approach for the treatment of patients carrying mutant FLT3. Activated FLT3 is constitutively phosphorylated on several tyrosine residues. These tyrosine residues facilitate association of SH2 domain-containing signaling proteins. By using a panel of SH2 domain-containing proteins we identified SLAP2 as a potent interaction partner of FLT3. The interaction in between FLT3 and SLAP2 occurs when FLT3 is activated and an intact SH2 domain of SLAP2 is required for the interaction. SLAP2 associates with FLT3 mainly through its SRC binding sites and expression of SLAP2 inhibited oncogenic FLT3-ITD-mediated cell proliferation and colony formation in vitro, and tumor formation in vivo. By analysis of patient expression data, we found that loss of SLAP2 expression correlates with poor prognosis of AML patients carrying FLT3-ITD. SLAP2 inhibits FLT3-mediated downstream signaling such as activation of AKT, ERK, p38 and STAT5. Inhibition is partially mediated through ubiquitination-mediated degradation of FLT3. Taken together our current study demonstrates that SLAP2 is an important regulator of FLT3-mediated oncogenic signaling and thus modulation of the SLAP2 expression levels can be an alternative approach for the treatment of FLT3-ITD positive malignancies. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 103 (4) ◽  
pp. 1398-1407 ◽  
Author(s):  
Parham Minoo ◽  
Maryam Mohsen Zadeh ◽  
Robert Rottapel ◽  
Jean-Jacques Lebrun ◽  
Suhad Ali

Abstract SHP-1, an src homology 2 (SH2) domain containing protein tyrosine phosphatase, functions as a negative regulator of signaling downstream of cytokine receptors, receptor tyrosine kinases and receptor complexes of the immune system. Dephosphorylation of receptors and/or receptor-associated kinases has been described as the mechanism for the function of SHP-1. Here we demonstrate a novel mechanism by which SHP-1 down-regulates the Janus kinase–2 (Jak2)/signal transducer and activator of transcription-5 (Stat5) pathway downstream of the prolactin receptor (PRLR) and the erythropoietin receptor (EPOR) in a catalytic activity–independent manner. Structural/functional analysis of SHP-1 defined the C-terminal tyrosine residues (Y278, Y303, Y538, Y566) within growth factor receptor–bound protein 2 (Grb-2) binding motif to be responsible for delivering the inhibitory effects. Our results further indicate that these tyrosine residues, via recruitment of the adaptor protein Grb-2, are required for targeting the inhibitory protein suppressor of cytokine signaling–1 (SOCS-1) to Jak2 kinase. Finally, loss of SOCS-1 expression in SOCS-1–/– mouse embryonic fibroblast (MEF) cells led to attenuation in SHP-1 function to down-regulate PRL-induced Stat5 activation. All together, our results indicate that SHP-1 inhibits PRLR and EPOR signaling by recruitment and targeting of SOCS-1 to Jak2, highlighting a new mechanism of SHP-1 regulation of cytokine-receptor signaling.


2021 ◽  
Author(s):  
Neda Sadat Kazemein Jasemi ◽  
Christian Herrmann ◽  
Eva Magdalena Estirado ◽  
Lothar Gremer ◽  
Dieter Willbold ◽  
...  

Growth factor receptor-bound protein 2 (GRB2) is a trivalent adaptor protein and a key element in signal transduction. It interacts via its flanking nSH3 and cSH3 domains with the proline-rich domain (PRD) of the RAS activator SOS1 and via its central SH2 domain with phosphorylated tyrosine residues of receptor tyrosine kinases (RTKs; e.g., HER2). The elucidation of structural organization and mechanistic insights into GRB2 interactions, however, remain challenging due to their inherent flexibility. This study represents an important advance in our mechanistic understanding of how GRB2 links RTKs to SOS1. Accordingly, it can be proposed that (1) HER2 pYP-bound SH2 potentiates GRB2 SH3 domain interactions with SOS1 (an allosteric mechanism); (2) the SH2 domain blocks cSH3,enabling nSH3 to bind SOS1 first before cSH3 follows (an avidity-based mechanism); and (3) the allosteric behavior of cSH3 to other domains appears to be unidirectional, although there is an allosteric effect between the SH2 and SH3 domains.


1999 ◽  
Vol 189 (8) ◽  
pp. 1181-1194 ◽  
Author(s):  
Anne Marie-Cardine ◽  
Henning Kirchgessner ◽  
Eddy Bruyns ◽  
Andrej Shevchenko ◽  
Matthias Mann ◽  
...  

T lymphocytes express several low molecular weight transmembrane adaptor proteins that recruit src homology (SH)2 domain–containing intracellular molecules to the cell membrane via tyrosine-based signaling motifs. We describe here a novel molecule of this group termed SIT (SHP2 interacting transmembrane adaptor protein). SIT is a disulfide-linked homodimeric glycoprotein that is expressed in lymphocytes. After tyrosine phosphorylation by src and possibly syk protein tyrosine kinases SIT recruits the SH2 domain–containing tyrosine phosphatase SHP2 via an immunoreceptor tyrosine-based inhibition motif. Overexpression of SIT in Jurkat cells downmodulates T cell receptor– and phytohemagglutinin-mediated activation of the nuclear factor of activated T cells (NF-AT) by interfering with signaling processes that are probably located upstream of activation of phospholipase C. However, binding of SHP2 to SIT is not required for inhibition of NF-AT induction, suggesting that SIT not only regulates NF-AT activity but also controls NF-AT unrelated pathways of T cell activation involving SHP2.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3733-3733
Author(s):  
Gordon Chan ◽  
Demetrios Kalaitzidis ◽  
Tatiana Usenko ◽  
Jeffery Kutok ◽  
Wentian Yang ◽  
...  

Abstract The Src homology-2 (SH2) domain-containing phosphatase 2 (Shp2), encoded by Ptpn11, is an ubiquitously expressed non-receptor protein tyrosine phosphatase (PTP) that positively regulates Ras/Erk activation in many receptor protein tyrosine kinases (RTK) and cytokine receptors. Mutations in PTPN11 are found in ~35% of patients with juvenile myelomonocytic leukemia (JMML) and at lower incidence in other neoplasms. To model JMML pathogenesis, we generated knock-in mice that conditionally express the leukemia-associated mutant Ptpn11D61Y. Ptpn11D61Y expression in all hematopoietic cells evokes a fatal myeloproliferative disorder (MPD), featuring leukocytosis, anemia, hepatosplenomegaly with extramedullary hematopoiesis, and factor-independent colony formation by bone marrow (BM) and spleen cells. The Lin−Sca1+cKit+ (LSK) compartment is expanded and “right-shifted”, accompanied by increased stem cell factor (SCF)-evoked colony formation and Erk and Akt activation. However, stem cell activity is decreased in diseased mice, and mice engrafted with Ptpn11D16Y stem cells fail to develop MPD. Ptpn11D16Y common myeloid (CMP) and granulocyte-monocyte (GMP) progenitors produce cytokine-independent colonies in a cell-autonomous manner, and demonstrate elevated Erk and Stat5 activation in response to granulocyte-macrophage colony-stimulating factor (GM-SCF) stimulation. Ptpn11D61Y megakaryocyte-erythrocyte progenitors (MEP) yield increased numbers of erythrocyte burst-forming units (BFU-E), but MEP and erythrocyte-committed progenitors (EP) produce fewer erythrocyte colonyforming units (CFU-E), indicating defective erythroid differentiation. Our studies provide a mouse model for Ptpn11-evoked MPD and show that this disease results from cellautonomous, and distinct lineage-specific effects of mutant Ptpn11 on multiple stages of hematopoiesis.


Sign in / Sign up

Export Citation Format

Share Document