mutation of thr466 in shp2 abolishes its phosphatase activity, but provides a new substrate-trapping...

12
Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant Rebecca Merritt a , Michael J. Hayman b , Yehenew M. Agazie a, a Department of Biochemistry and Molecular Pharmacology and The Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506-9142, USA b Department of Molecular Genetics and Microbiology, Health Sciences Center, Stony Brook University, Stony Brook, New York, 11794-5222, USA Received 19 April 2005; received in revised form 8 November 2005; accepted 11 November 2005 Available online 20 Decembre 2005 Abstract Most classical phosphotyrosyl phosphatases (PTPs), including the Src homology phosphotyrosyl phosphatase 2 (SHP2) possess a Thr or a Ser residue immediately C-terminal to the invariant Arg in the active site consensus motif (H/V-C-X 5 -R-S/T), also known as the signature motif. SHP2 has a Thr (Thr466) at this position, but its importance in catalysis has not been investigated. By employing site-directed mutagenesis, phosphatase assays and substrate-trapping studies, we demonstrate that Thr466 is critical for the catalytic activity of SHP2. Its mutation to Ala abolishes phosphatase activity, but provides a new substrate-trapping mutant. We further show that the nucleophilic Cys459 is not involved in substrate trapping by Thr466Ala-SHP2 (T/A-SHP2). Mutation of Thr466 does not cause significant structural changes in the active site as revealed by the trapping of the epidermal growth factor receptor (EGFR), the physiological substrate of SHP2, and by orthovanadate competition experiments. Based on these results and previous other works, we propose that the role of Thr466 in the catalytic process of SHP2 could be stabilizing the sulfhydryl group of Cys459 in its reduced state, a state that enables nucleophilic attack on the phosphate moiety of the substrate. The T/A-SHP2 harbors a single mutation and specifically interacts with the EGFR. Since the nucleophilic Cys459 and the proton donor Asp425 are intact in the T/A-SAHP2, it offers an excellent starting material for solving the structure of SHP2 in complex with its physiological substrate. © 2005 Elsevier B.V. All rights reserved. Keywords: SHP2; Phosphatase; EGFR; Substrate trapping 1. Introduction Protein Tyr phosphorylation serves as one of the mechanisms for signal transduction induced by the binding of growth factors, cytokines or the extracellular matrix to cognate receptors. It has become evident that dephosphorylation reactions are as important as phosphorylation reactions. The enzymes that catalyze phosphorylation reactions are either the receptors themselves with intrinsic tyrosine kinase activity, known as receptor tyrosine kinases (RTKs) or cytoplasmic tyrosine kinases that are recruited to the plasma membrane upon stimulation [13]. On the other hand, dephosphorylation reactions are catalyzed by a family of enzymes called phosphotyrosyl phosphatases (PTPs). PTPs are classified as transmembrane or cytoplasmic based on their localization within the cell under basal conditions [4]. Alternatively, they are classified as classical PTPs, those specific to Tyr(P) or dual- specificity PTPs, those that catalyze Tyr(P), Thr(P) and Ser(P) dephosphorylation reactions [46]. The mammalian Src homology 2 phosphotyrosyl phospha- tase 1 and 2 (SHP1 and SHP2) belong to the cytoplasmic classical PTPs [7]. The identifying feature of SHP1 and SHP2 is the presence of two tandemly-arranged SH2 domains in their N-terminal region [8,9]. SHP2 is a ubiquitously expressed mammalian PTP, whereas SHP1 is primarily restricted to hematopoietic cells [6,911]. SHP1 and SHP2 show significant structural homology both in the SH2 and PTP domains, but play opposite physiological roles. SHP1 is a negative regulator of tyrosine kinase signaling, whereas SHP2 is primarily a positive effector [4,6,12]. The Drosophila melanogaster PTP, Cork- screw, also is an SH2 domain-containing PTP with functional Biochimica et Biophysica Acta 1763 (2006) 45 56 http://www.elsevier.com/locate/bba Corresponding author. Tel.: +1 304 293 7756; fax: +1 304 293 6846. E-mail address: [email protected] (Y.M. Agazie). 0167-4889/$ - see front matter © 2005 Elsevier B.V. All rights reserved. doi:10.1016/j.bbamcr.2005.11.013

Upload: rebecca-merritt

Post on 04-Sep-2016

221 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

ta 1763 (2006) 45–56http://www.elsevier.com/locate/bba

Biochimica et Biophysica Ac

Mutation of Thr466 in SHP2 abolishes its phosphatase activity,but provides a new substrate-trapping mutant

Rebecca Merritt a, Michael J. Hayman b, Yehenew M. Agazie a,⁎

a Department of Biochemistry and Molecular Pharmacology and The Robert C. Byrd Health Sciences Center,West Virginia University, Morgantown, WV 26506-9142, USA

b Department of Molecular Genetics and Microbiology, Health Sciences Center, Stony Brook University, Stony Brook, New York, 11794-5222, USA

Received 19 April 2005; received in revised form 8 November 2005; accepted 11 November 2005Available online 20 Decembre 2005

Abstract

Most classical phosphotyrosyl phosphatases (PTPs), including the Src homology phosphotyrosyl phosphatase 2 (SHP2) possess a Thr or a Serresidue immediately C-terminal to the invariant Arg in the active site consensus motif (H/V-C-X5-R-S/T), also known as the “signature motif”.SHP2 has a Thr (Thr466) at this position, but its importance in catalysis has not been investigated. By employing site-directed mutagenesis,phosphatase assays and substrate-trapping studies, we demonstrate that Thr466 is critical for the catalytic activity of SHP2. Its mutation to Alaabolishes phosphatase activity, but provides a new substrate-trapping mutant. We further show that the nucleophilic Cys459 is not involved insubstrate trapping by Thr466Ala-SHP2 (T/A-SHP2). Mutation of Thr466 does not cause significant structural changes in the active site as revealedby the trapping of the epidermal growth factor receptor (EGFR), the physiological substrate of SHP2, and by orthovanadate competitionexperiments. Based on these results and previous other works, we propose that the role of Thr466 in the catalytic process of SHP2 could bestabilizing the sulfhydryl group of Cys459 in its reduced state, a state that enables nucleophilic attack on the phosphate moiety of the substrate.The T/A-SHP2 harbors a single mutation and specifically interacts with the EGFR. Since the nucleophilic Cys459 and the proton donor Asp425are intact in the T/A-SAHP2, it offers an excellent starting material for solving the structure of SHP2 in complex with its physiological substrate.© 2005 Elsevier B.V. All rights reserved.

Keywords: SHP2; Phosphatase; EGFR; Substrate trapping

1. Introduction

Protein Tyr phosphorylation serves as one of the mechanismsfor signal transduction induced by the binding of growthfactors, cytokines or the extracellular matrix to cognatereceptors. It has become evident that dephosphorylationreactions are as important as phosphorylation reactions. Theenzymes that catalyze phosphorylation reactions are either thereceptors themselves with intrinsic tyrosine kinase activity,known as receptor tyrosine kinases (RTKs) or cytoplasmictyrosine kinases that are recruited to the plasma membrane uponstimulation [1–3]. On the other hand, dephosphorylationreactions are catalyzed by a family of enzymes calledphosphotyrosyl phosphatases (PTPs). PTPs are classified as

⁎ Corresponding author. Tel.: +1 304 293 7756; fax: +1 304 293 6846.E-mail address: [email protected] (Y.M. Agazie).

0167-4889/$ - see front matter © 2005 Elsevier B.V. All rights reserved.doi:10.1016/j.bbamcr.2005.11.013

transmembrane or cytoplasmic based on their localizationwithin the cell under basal conditions [4]. Alternatively, they areclassified as classical PTPs, those specific to Tyr(P) or dual-specificity PTPs, those that catalyze Tyr(P), Thr(P) and Ser(P)dephosphorylation reactions [4–6].

The mammalian Src homology 2 phosphotyrosyl phospha-tase 1 and 2 (SHP1 and SHP2) belong to the cytoplasmicclassical PTPs [7]. The identifying feature of SHP1 and SHP2is the presence of two tandemly-arranged SH2 domains in theirN-terminal region [8,9]. SHP2 is a ubiquitously expressedmammalian PTP, whereas SHP1 is primarily restricted tohematopoietic cells [6,9–11]. SHP1 and SHP2 show significantstructural homology both in the SH2 and PTP domains, but playopposite physiological roles. SHP1 is a negative regulator oftyrosine kinase signaling, whereas SHP2 is primarily a positiveeffector [4,6,12]. The Drosophila melanogaster PTP, Cork-screw, also is an SH2 domain-containing PTP with functional

Page 2: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

Fig. 1. The active site consensus motif or signature motif of six classical PTPs isshown. SHP2, SHP1, Corkscrew or R-PTPα has a Thr (T), while PTP1B orVHR has a Ser (S) residue immediately C-terminal to the invariant Arg (R). Inthe signature motif, Cys (C) acts as a nucleophile and R as a phosphatecoordinator.

46 R. Merritt et al. / Biochimica et Biophysica Acta 1763 (2006) 45–56

similarity to SHP2 [13–15]. Several lines of evidence show thatSHP2 is important for normal growth and development [16–18]. However, the molecular mechanism of SHP2 in varioussignaling pathways is poorly understood. One of the majorstumbling blocks in SHP2 studies has been that its physiologicalsubstrates have not been identified. To overcome this hurdle, asubstrate-trapping mutant of SHP2 termed DM-SHP2 (DM forD425A/C459S double mutant) was recently developed [19].Using this substrate-trapping mutant a molecular mechanism forSHP2 in the epidermal growth factor receptor (EGFR) signalingpathway has been described [19].

SHP2 possesses the common structural features found in thePTP domains of all classical PTPs- the active site consensusmotif (also called the signature motif) identified as H/V–C–X5–R–S/T and the W–P–D loop [4,20]. Currently, it is believed thatmost PTPs follow the same catalytic principle in depho-sphorylating Tyr(P) on target substrates. The same could be truefor SHP2 given the presence of all the necessary residues forphosphatase (PTPase) activity. Upon binding of a substrate tothe active site, the invariant Cys residue makes a nucleophilicattack on the phosphoester bond. This leads to the cleavage ofthe phosphate moiety from the target Tyr residue and theformation of a cysteinyl–phosphate intermediate [20]. The Aspresidue in the W–P–D loop donates a proton to satisfy therequirements for the leaving phenolate group. The deprotonatedAsp then acts as a general base and activates a water molecule,which will induce the hydrolysis of the cysteinyl–phosphateintermediate [21]. In addition, the conserved Gln residue C-terminal to the signature motif has been proposed to be involvedin the hydrolysis of the cysteinyl–phosphate intermediate [22].The invariant Arg residue mediates substrate binding into theactive site by coordinating the phosphate moiety. Recently, itwas demonstrated that substitution of Arg465 with Gluabrogates the substrate binding ability of SHP2 and rendersthe enzyme inactive [19] confirming the universal role of Arg inthe signature motif of PTPs.

Based on the above catalytic principle, substrate-trappingmutants of several PTPs have been produced by mutation of theW–P–D loop Asp to Ala [23,24]. In contrast, a correspondingmutation in SHP2 (Asp425Ala) does not provide a substrate-trapping mutant [19,25]. Although these results cannot rule outthe role of Asp425 in the catalytic process of SHP2, theydefinitely indicate the presence of additional players, which arenot yet defined. All classical PTPs, including SHP2, possess aThr or a Ser immediately C-terminal to the invariant Argresidue; SHP2 has a Thr at this position (Thr466) [4,26–30].The same is true for SHP1, Corkscrew and R-PTPα. On theother hand PTP1B and vaccinia H1-related (VHR) have a Serresidue at this position (Fig. 1). In VHR, it was suggested thatthe Ser residue mediates the hydrolysis of a cysteinyl–phosphate intermediate during the dephosphorylation reaction[31]. In the current report, we set out to determine theimportance of The466 in the catalytic process SHP2. Byemploying site-directed mutagenesis, substrate-trapping studiesand PTPase assays, we demonstrate that mutation of Thr466 toAla abolishes the PTPase activity of SHP2, but provides a newsubstrate trapping mutant, which is as efficient as the DM-SHP2

previously reported [19]. In addition, this mutation does notaffect substrate specificity as revealed by the trapping of theepidermal growth factor receptor (EGFR), the physiologicalsubstrate of SHP2.

2. Materials and methods

2.1. Cells, cell culture, antibodies, etc.

The cell types used in this study were COS-1 and NIH-3T3. All cell typeswere grown in Dulbecco's Modified Eagle's Medium (DMEM) supplementedwith 10% foetal calf serum (FCS), and maintained at 37 °C with 7% CO2. TheEpidermal growth factor (EGF) was purchased from GIBCO-BRL, anti-EGFRmonoclonal antibody from Upstate Biotechnologies, anti-PTP1D (SHP2) fromPharmagen, and anti-Tyr(P) monoclonal antibody from Cell Signaling. Thepolyclonal antibody against SHP2 was raised by injection of rabbits with GSTfusion of the N-terminal region [32]. Horseradish peroxidase-conjugatedsecondary antibodies were purchased from Amersham.

2.2. Plasmid construction and site-directed mutagenesis

The construction of plasmids that express WT-SHP2, Asp425Ala-SHP2,Cys459Ser-SHP2 and Asp425Ala/Cys459Ser-SHP2 was described previously[19,32]. Hereinafter, these mutants are referred to asWT-SHP2, D/A-SHP2, C/S-SHP2 and DM-SHP2 (DM for double mutant), respectively. T466A-SHP2 andC459S/T466A-SHP2mutants hereinafter referred to as the T/A-SHP2 and the C/S-T/A-SHP2, respectively, were produced by introducing the indicated pointmutation in the WT- or the C/S-SHP2 background using the following primersthat span the signature motif. The sense primer was 5′-CTGCAGTGCTG-GAATTGGCCGG-GCAGGGACGTTCATTGTG-3′ and the anti-sense primerwas 5′-CACAATGAACGTCCCTGCCCGGCCAATTCCAGC-ACTGCAG-3′.The Stratagene site-directed mutagenesis kit and protocol was used to producethese mutants. Introduction of the point mutation was confirmed by sequencingthe relevant region. The glutathione S-transferase fusion of the PTP domain of theWT-, the T/A-, the C/S-T/A- or the DM-SHP2 protein was constructed by PCRamplification of the corresponding PTP domains, and ligation into the pGEX-KGvector at BamHI and HindIII sites. The sense primer containing BamHIrestriction endonuclease site was 5′-AATTTTGGATCCCCCCTTAACAC-GACTCGTATAAATG-3′, and the anti-sense primer containing the HindIIIsite was 5′-AAATTTTAAGCTTTCATCTGAAACTTTTC-TGCTGTTGCAT-C-3′. These fusion proteinswere designated asWT-PTP, T/A-PTP, C/S-T/A-PTP,and DM-PTP.

2.3. Cell transfection and preparation of lysates

Cells were transfected with various SHP2 constructs using the FuGenetransfection regent as recommended by the manufacturer (Roche). Followingtransfection, cells were incubated for ∼36 h, serum-starved for ∼12 h and thenstimulated with 100 ng/ml EGF. After washing twice with ice-cold PBS, cellswere lysed in 20 mM Tris–HCl pH 7.4 buffer containing 150 mMNaCl, 50 mMNaF, 1 mMEDTA, 10% glycerol, 1% triton-X-100, 1 mMNa orthovanadate and

Page 3: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

47R. Merritt et al. / Biochimica et Biophysica Acta 1763 (2006) 45–56

protease inhibitor cocktail (Roche). Lysates were cleared by centrifugation at15,000×g at 4 °C and analyzed as desired.

2.4. PTPase assay

The PTPase activity of the various GST fusion proteins was performed withslight modification of the previously described protocol [31,33]. Briefly, GST-PTPs were purified on glutathione sepharose beads as recommended by themanufacturer (Pharmacia), and quantified by measuring absorbance at 280 nmand comparison with BSA standards. Para-nitrophenyl phosphate (pNPP)(Biomol) was used as an artificial substrate to determine the PTPase activity ofeach GST-PTP. Reactions were performed at 30 °C in 100 μl volume of PTPasebuffer containing 50 mM sodium acetate (for pHs 3.0–5.5) or 25 mM Tris–HCl(for pHs 6.0–9.0), 50 mM NaCl, 2 mM EDTA and 10 mM dithiothreitol (DTT).Change in absorbance was followed in a plate-reading visible spectrophotometer(TECAN) at 405 nm.

2.5. Immunoprecipitation and immunoblotting

Cleared cell lysates were incubated with anti-SHP2 polyclonal antibodyovernight at 4 °C with an additional incubation for 1 h after addition of protein-Asepharose beads. The precipitates were washed 3 times with cell lysis buffer,eluted by boiling with Laemmli sample buffer, and then separated on 10%denaturing polyacrylamide gel. After transfer onto nitrocellulose membrane,blocking was done by incubation with 3% bovine serum albumin. Membraneswere then stained with the primary antibody either overnight at 4 °C or for 2 h atroom temperature. The secondary antibody staining was for 1 h at roomtemperature. The chemiluminescence detection method was used for all Westernblot experiments.

2.6. Affinity precipitation studies

The GST-fusion of the PTP domains of the various SHP2 mutants wereprepared as described previously [19,34]. For affinity precipitation, GST fusionproteins were first captured on glutathione sepharose beads by incubation ofcleared bacterial lysates (∼1 μg GST-PTP) for 30 min at 4 °C [19,34]. Capturedproteins were washed three times with bacterial lysis buffer and two times witheukaryotic cell lysis buffer. Cell lysates from COS-1 or NIH-3T3 were thenadded to beads and incubated for 1 h at 4 °C, washed five times with cell lysisbuffer, and eluted by boiling for 10 min with Laemmli sample buffer. Separationof proteins and immunoblot analysis was as described above.

3. Results

Most classical PTPs possess a conserved Ser or Thr residueimmediately C-terminal to the invariant Arg in the active sitesignature motif (Fig. 1) [4,26–30]. In the dual specificityphosphatase VHR (for vaccinia H1-Related), the Ser residue inthis position (Ser131) was shown to be important for enzymeturnover [31]. SHP2 has a Thr at this position (Thr466), but itsrelevance in the catalytic process of this enzyme has not beenaddressed. Thus, we investigated the importance of Thr466 inthe catalytic process of SHP2 by a combination of in vitroPTPase assays, site-directed mutagenesis and substrate-bindingstudies in the presence or absence of PTP inhibitors.

3.1. The T/A-SHP2 protein is a PTPase-dead mutant

T/A-SHP2 was produced by site-directed mutagenesis asdescribed in the materials and methods. The consequence ofthis mutation on the PTPase activity of SHP2 was tested in twoways—by determining its state of Tyr phosphorylationfollowing EGF stimulation and by in vitro PTPase assays.

SHP2 becomes phosphorylated on Tyr542 and Tyr580 uponstimulation of cells with growth factors or serum [13,35,36],and as a mechanism of autoregulation, it dephosphorylates itselfimmediately [37]. We used this phenomenon as an initial testfor the PTPase activity of the T/A-SHP2. The DM-SHP2 [19]and the C/S-SHP2 [38,39] were used as PTPase-dead controls,while the WT-SHP2 and the D/A-SHP2 as positive controls.These constructs were transiently expressed in COS-1 cells andEGF-induced Tyr phosphorylation and autodephosphorylationwere determined by immunoblotting with anti-Tyr(P) antibody.The T/A-SHP2, like those devoid of the nucleophilic Cys459(C/S-SHP2 or DM-SHP2), was highly Tyr phosphorylated inEGF-stimulation-dependent manner (Fig. 2A, top panel). Asreported previously, the Tyr phosphorylation of the D/A-SHP2mutant was similar to that of the WT-SHP2 protein [19].Moreover, the Tyr phosphorylation state of the EGFR, the onlycharacterized SHP2 substrate, and the three other potentialsubstrates previously identified as p90, p120 and p150, washigher in the T/A-, the C/S- and the DM-SHP2 cells. In fact, thephosphorylation of p90 and p150 was undetectable in vector,WT-SHP2 and D/A-SHP2 cells. Reblotting the membrane withanti-EGFR antibody showed that there was no difference in theamount of this protein loaded to each lane (Fig. 2A, middlepanel). Further reblotting with anti-SHP2 antibody showed thatthe expression of the different SHP2 proteins was comparable(Fig. 2A, bottom panel). These results suggest that the T/A-SHP2 could be PTPase defective.

Because autodephosphorylation alone is insufficient toconclude that the T/A-SHP2 protein is inactive, we conductingin vitro PTPase assays. We used purified PTP domains (GSTfusions) of the WT- or the T/A-SHP2 protein as an enzyme andpara-nitrophenyl phosphate (pNPP) as an artificial substrate(see materials and methods). First, PTPase assays wereconducted at different pH points in the presence of 1 μg PTPand a saturating concentration of pNPP (100 mM) to see if theT/A-PTP and WT-PTP have different pH preferences oroptimum points. For each pH point, reactions containingpNPP only were used as negative controls. The reactionswere followed with absorbance measurement (OD) at 405 nm ina plate reader spectrophotometer (TECAN). Absorbance at 5min time point was used to compare the effect of pH on enzymeactivity. As shown in Fig. 2B, there was no detectable activity inthe T/A-PTP and control samples at any pH points tested. Onthe other hand, the WT-PTP showed a sharp increase inabsorbance at pH 5.0, reached maximum at pH 7.0, anddeclined sharply after that, providing a typical bell-shaped curvewith narrow range. To test if time could be a factor on thePTPase activity T/A-PTP, the absorbance values at pH 7.0 wereplotted against time. Again, the T/A-PTP did not show anyactivity at all time points tested, while the WT-PTP exhibited asteady increase with time (Fig. 2C). Further kinetic analysiscould not be conducted simply because the T/A-PTP wasenzymatically inactive. These results demonstrate that Thr466 isrequired for the catalytic activity of SHP2. Furthermore, theydemonstrate that maximum PTPase activity of SHP2 occurs atpH 7.0, a pH that corresponds to the cytosolic milieu. Given thecomplete loss of PTPase activity, the role of Thr466 in SHP2

Page 4: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

Fig. 2. Effect of Thr466Ala mutation on the PTPase activity of SHP2. (A) Theindicated SHP2 proteins were transfected into COS-1 cells. After incubation for∼36 h in growth medium and ∼12 h in serum-free medium, cells werestimulated with 100 ng/ml EGF for 10 min and lysed. Total cell lysates wereseparated by SDS-PAGE and analyzed by immunoblot with the indicatedantibodies. Note that the C/S-the T/A- or the DM-SHP2 mutant is hypertyrosylphosphorylated. PTPase assays using para-nitrophenol phosphate (pNPP) as asubstrate at different pH points (B) or time points (C) are depicted in graphs. ThePTPase activity of the WT-PTP or T/A-PTP was determined by measuringabsorbance (production of para-nitrophenol) at 405 nm in a plate readerspectrophotometer. Three independent experiments were performed in eachcase, and the results were plotted absorbance versus pH in B or absorbanceversus time (min) in C.

48 R. Merritt et al. / Biochimica et Biophysica Acta 1763 (2006) 45–56

may not be mediating the hydrolysis of a cysteinyl phosphateintermediate as proposed for VHR.

3.2. T/A-SHP2 is a substrate-trapping mutant

As demonstrated above, the T/A-SHP2 protein is a PTPase-dead mutant. Loss of PTPase activity could emanate from loss ofsubstrate binding or inability to hydrolyze the phosphodiesterbond. To gain some insight on this point, we tested the ability ofthe T/A-SHP2 to bind to the physiological substrate of SHP2, theEGFR. Lysates prepared as in Fig. 2Awere immunoprecipitatedwith anti-SHP2 antibody and analyzed by immunoblot with theindicated antibodies. Similar to the DM-SHP2 protein reportedpreviously [19], the T/A-SHP2 protein trapped p170, whichrepresents the EGFR, and three other unknown phosphotyrosylproteins identified as p150, p120 and p90 (Fig. 3A, top). TheC/S-SHP2 also trapped the EGFR, p150 and p120, but ascompared to the T/A- or the DM-SHP2, it was inefficient. Itsinefficiency was more apparent when it failed to trap the less-abundant p90. On the other hand, the WT- or the D/A-SHP2protein exhibited a much lower binding to p170 (EGFR), but notothers. Again, the tyrosine phosphorylation of p70, whichrepresents SHP2, was higher in the T/A-, the C/S- and the DM-SHP2 proteins. The identity of p170 and p70 was confirmed byreblotting the same membrane with anti-EGFR and anti-SHP2antibodies (Fig. 3A, middle and bottom panels, respectively).The anti-EGFR monoclonal antibody blot that recognizes asingle epitope better demonstrates the trapping efficiency of theT/A- or the DM-SHP2 and the inefficiency of the C/S-SHP2than the anti-Tyr(P), which recognizes multiple epitopes(compare top and middle panels, Fig. 3A). These results suggestthat although the T/A-SHP2 protein is a PTPase-dead mutant, itretains its substrate-binding property. Given the similarity insubstrate binding efficiency between the T/A-SHP2 and DM-SHP2, it is highly likely that the T/A-SHP2 is another substrate-trapping mutant. In conformity with our previous report [19], theD/A-SHP2 protein behaved like the WT-SHP2 in its interactionwith the EGFR and in its level of Tyr phosphorylation.

Substrate trapping refers to a stable interaction between atarget substrate and a mutant PTP domain. The results presentedin Fig. 3A show that some amount of the EGFR protein wascoprecipitated with the WT- or the D/A-SHP2 protein. Thiscould be due to SH2 domain-mediated interactions since the PTPdomains of these proteins are PTPase active, and thus cannot trapsubstrates. To confirm this assumption, the PTP domains of theindicated SHP2 proteins were produced as GST-fusions (seematerials andmethods) and used to affinity precipitate the EGFRfrom COS-1 cell lysates stimulated with EGF. As expected, theEGFR was affinity precipitated by the T/A-PTP and the DM-PTP. The C/S-PTP also precipitated the EGFR, but whencompared with the T/A-PTP or DM-PTP, it was significantlylower. On the other hand, there was basal precipitation by WT-PTP or the D/A-PTP and negligible by the GST alone (Fig. 3B,top). Reblotting with anti-Tyr(P) antibody showed that the threeother as yet unidentified phosphotyrosyl proteins also wereaffinity precipitated by T/A- or DM-PTP suggesting that theydirectly interact with the PTP domain (Fig. 3B, middle panel).

Page 5: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

Fig. 3. Substrate binding studies. A) COS-1 cells were transfected with theindicated SHP2 proteins, incubated for 36 h, serum starved for 12 h, and thenstimulated with 100 ng/ml EGF. Lysates prepared from these cells were subjectedto immunoprecipitation (IP) with anti-SHP2 and immunoblotting (IB) with anti-Tyr(P) (top panel), anti-EGFR (middle panel) or anti-SHP2 (bottom panel)antibody. (B) Affinity precipitation studies. COS-1 cells were grown toconfluency, serum starved for 12 h and then stimulated with EGF for 10 min.Lysates prepared from these cells were affinity precipitated (AP) with theindicated GST fusions of the PTP domains. The precipitates were then analyzedby immunoblotting with anti-EGFR (top panel), anti-Tyr(P) antibody (middlepanel) or anti-GST (bottom panel) antibody. Band densities of EGFR precipitatedwith the different GST fusion protein is shown at the top of the top panel.

49R. Merritt et al. / Biochimica et Biophysica Acta 1763 (2006) 45–56

Further reblotting the membranes with anti-GST antibodyconfirmed that the amount of all PTP domains used wascomparable (Fig. 3B, bottom panel). These results confirm thatthe coprecipitation of EGFR with the WT- or the D/A-SHP2proteins was due to interaction via the SH2 domains. Therefore,the T/A-SHP2 protein represents a new substrate-trappingmutant.

3.3. Effect of orthovanadate on substrate trapping

Orthovanadate (OV) competitively inhibits PTPs by bindingto the active site of these enzymes. As a result, it is routinely

used as an inhibitor of PTPs in both in vitro and in vivo studies[40,41]. To evaluate whether this protocol could fit to ourexperimental conditions, we first tested the efficiency of OV inincreasing Tyr phosphorylation of cellular proteins in intactcells. We treated serum-starved COS-1 cells with varyingconcentration of OV for 30 min and performed anti-Tyr(P) blotanalysis of total cell lysates. As shown in Fig. 4A, OV increasedTyr phosphorylation in a concentration-dependent manner inthe absence of EGF stimulation. Thus, it does cross the PM andinhibit PTPs. Anti-actin reblotting showed that there was nodifference in the amount of total protein loaded to each lane.Next, we investigated the effect of OVon substrate trapping bySHP2 proteins. COS-1 cells transiently expressing WT-SHP2,T/A-SHP2 or DM-SHP2 proteins were serum starved, treatedwith OVand then stimulated with EGF for 10 min. Samples nottreated with OV or EGF were also included as controls. Todetermine pattern of Tyr phosphorylation under these condi-tions, total cell lysates were analyzed by immunoblotting.Clearly, addition of OV over EGF provides an increase innumber and intensity of phosphorylated proteins even in cellsoverexpressing WT-SHP2 (Fig. 4B, top panel). Reblotting withanti-SHP2 antibody indicated that all constructs were expressedequally (Fig. 4B, bottom panel). These lysates were subjected toimmunoprecipitation with anti-SHP2 and immunoblotting withanti-EGFR antibodies. OV did not affect the trapping of theEGFR by the T/A- or the DM-SHP2 protein up to 5 mM, butinhibited by approximately 50% at 10 mM (Fig. 4C, top panel).The interaction of the WT-SHP2 protein with the EGFR wasunaffected by OV treatment suggesting that it was solely SH2domain-mediated. Reblotting with anti-SHP2 antibody showedthat amount of SHP2 proteins was comparable (Fig. 4C, lowerpanel). These results show that OV does not affect substratetrapping by the T/A- or the DM-SHP2 proteins at lowerconcentrations, but could inhibit if used higher than 5 mM inCOS-1 cells.

The above findings raised an important question of whymore than 5 mM OV was needed to induce an inhibitory effecton substrate trapping. Although we observed a direct relation-ship between OV concentration and protein Tyr phosphoryla-tion, it was difficult to determine how much of extracellularlyadded OV could in fact cross the cell membrane. We did notquantify the concentration of the EGFR (the test substrate) inour lysates, but we predicted that it was much lower than the OVused. If that was the case, it could either be that the EGFR has amuch higher affinity for binding to the active site or that theamount of OV that crossed the plasma membrane and competedwith the EGFR was significantly lower than the amount added.This prompted us to conduct substrate trapping studies outsidethe cell where we can control the concentration of OV. For theseexperiments, COS-1 cells were serum starved for ∼12 h, leftuntreated or treated with 1 mM OV for 30 min (to protectphosphorylation sites on EGFR), stimulated with EGF for 10min and lysed. These lysates were incubated with the GSTfusion of the PTP domain of the WT-, the T/A- or the DM-SHP2protein in the absence or presence of varying concentration ofOV. The precipitates were analyzed by immunoblotting withanti-EGFR antibody. As expected, the T/A- and DM-PTP

Page 6: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

Fig. 4. Orthovanadate treatment studies. (A) Serum-starved COS-1 cells were treated with the indicated amounts of orthovanadate (OV) for 30 min. Lysates prep d from these cells were analyzed by immunoblottingwith anti-Tyr(P) antibody (top panel). An anti-actin blot is included as a loading control (bottom panel). (B) The indicated SHP2 constructs were transiently exp ed in COS-1 cells as described in Fig. 2. After serumstarvation, cells were left untreated or treated with the indicated amounts of OV for 30 min by adding in the medium. They were then left unstimulated or stimula with EGF for 10 min. Total cell lysates prepared fromthese cells were analyzed for effect of OV on Tyr phosphorylation of cellular proteins and expression of the different SHP2 proteins. (C) The lysates in B wer bjected to immunoprecipitation with anti-SHP2 andimmunoblot analysis with anti-EGFR antibodies to determine OV's effect on substrate trapping. (D) Effect of OVon affinity precipitation of the EGFR by the T fusion of the PTP domains of the indicated SHP2proteins out side the cell. Numbers above the EGFR blot are band densities to better depict effect of OV.

50R.Merritt

etal.

/Biochim

icaet

Biophysica

Acta

1763(2006)

45–56

areresstede suGS

Page 7: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

51R. Merritt et al. / Biochimica et Biophysica Acta 1763 (2006) 45–56

trapped the EGFR, while the WT-PTP did not. Based on bandintensity measurements, the trapping was unaffected at 1 mM.However, it was 8 and 4 fold over basal at 2 and 5 mM,respectively, and only basal at 10 mM (Fig. 4D, top panel).Reblotting the membrane with anti-GST antibody showed thatthere was no difference in the amount of GST fusion proteins ineach lane (Fig. 4D, lower panel). Therefore, OV competitivelyinhibits substrate trapping by the T/A- or the DM-SHP2 protein.Furthermore, these results suggest that mutation of Th466 doesnot cause structural changes that interfere with substratebinding.

3.4. Mechanism of substrate trapping by the T/A-SHP2 protein

As demonstrated above, the T/A-SHP2 protein is a PTPase-dead, but a substrate-trapping mutant. The next logical step wasto investigate the role of Thr466 in the catalytic process ofSHP2. We addressed this question by site-directed mutagenesis,substrate binding and pervanadate treatment studies. In theactive site of SHP2 and other PTPs, the Cys residue acts as anucleophile, while the Arg as a phosphate coordinator. Mutationof the Cys residue to Ser in SHP2 provides a weaker substrate-trapping mutant that also acts as a dominant-negative protein[20,38,39], while mutation of Arg to Glu gives rise to adominant-negative protein with no substrate-binding property[42]. In other PTPs, mutation of the WPD loop Asp to Ala aloneprovides substrate trapping mutants [23,24,43], but not in SHP2[19]. Substrate trapping by the Asp-to-Ala mutants was shownto be due to accumulation of a cysteinyl phosphate intermediate[23,44], a covalently-linked intermediate between the thiolategroup of Cys and the phosphate group of the substrate. As aresult, these mutants show initial PTPase activity until allenzyme molecules are occupied. The same was true forSer131Ala mutant of VHR [31]. In case of T/A-SHP2, however,total loss of PTPase activity indicates that the role of Thr466might not be mediating the hydrolysis of a transition-statecomplex since no initial para-nitrophenol production wasobserved in the in vitro PTPase reactions (see Fig. 2B and C).Although these results argue against accumulation of a cysteinylphosphate intermediate, loss of enzymatic activity alone isinsufficient to exclude the involvement of Cys in substratetrapping by the T/A-SHP2. Therefore, we reasoned that specificinactivation of Cys459 in the background of T/A-SHP2 mightprovide insights on the mechanism of substrate trapping. Thus,we introduced a Cys459Ser mutation in the background of T/A-SHP2, hereinafter referred to as the C/S-T/A-SHP2 andconducted substrate-trapping studies. The T/A-, the C/S-T/A-or the DM-SHP2 protein was expressed in COS-1 cells, and theability of each protein to trap the EGFR was compared. TheWT-SHP2 protein was used as a negative control. Theseexperiments were conducted in time-course fashion to evaluatethe stability of enzyme-substrate complex following EGFstimulation. Initial analysis of total cell lysates showed thatthe T/A-, the C/S-T/A- and the DM-SHP2 proteins were highlyphosphorylated owing to their inability to autodephosphorylatethemselves (Fig. 5A, top panel). Reblotting with anti-SHP2antibody showed that amount of SHP2 protein in all lanes was

comparable (Fig. 5A, bottom panel). Immunoprecipitation withanti-SHP2 and immunoblotting with anti-EGFR antibodydemonstrated that the C/S-T/A-SHP2 protein trapped theEGFR as efficiently as the T/A- or the DM-SHP2 protein(Fig. 5B, top panel). In addition, the enzyme–substrate complexwas stable for at least 120 min following EGF stimulation. Onthe other hand, the WT-SHP2 protein showed low level ofinteraction that decreased over the 2-h stimulation period.Reblotting the membrane with anti-SHP2 antibody showed thatthere was no difference in the amount of each SHP2 proteinrecovered during the immunoprecipitation reaction (Fig. 5B,bottom panel). Thus, introduction of the Cys459Ser mutationinto the T/A-SHP2 did not affect the trapping propertysuggesting that Cys459 may not contribute to this phenomenon.However, these findings could potentially be complicated by thefact that the C/S-SHP2 protein by itself exhibits some level ofsubstrate trapping. To complement these findings, pervanadate(PV) treatment studies were conducted (see below).

Substrate trapping by the Asp-to-Ala mutants of other PTPswas proposed to be due to accumulation of a cysteinyl–phosphate intermediate [23,24,43]. As such, these mutantsexhibit sensitivity to the oxidative inhibitor pervanadate (PV)suggesting that the Cys residue is important for their trappingproperty [45,46]. Given the efficiency of the C/S-T/A-SHP2mutant, the presence of Cys459 did not seem to be importantfor trapping by the T/A-SHP2. However, it is possible that theT/A-SHP2 on one hand and the C/S-T/A-SHP2 on the othermay utilize different mechanisms to trap target substrateswhere Cys459 is necessary only for the T/A-SHP2. If thiswere the case, then treatment of cells with PV could abolishtrapping by the T/A-SHP2 due to oxidation of the sulfhydrylgroup. Initially, we analyzed the effect of PV on the Tyrphosphorylation of cellular proteins. As shown in Fig. 5C, PVenhanced Tyr phosphorylation in a concentration-dependentmanner similar to OV. Next, we tested PV's effect on substratetrapping by TA-SHP2 protein. The WT-SHP2 and DM-SHP2proteins were used as negative and positive controls,respectively. COS-1 cells were transfected with these con-structs and treated with varying concentration of PV for 30min prior to EGF stimulation. Analysis of total cell lysatesindicated that PV enhances Tyr phosphorylation under theseconditions too (Fig. 5D, top panel). Reblotting with anti-SHP2antibody showed that amount of overexpressed SHP2 proteinswas comparable (Fig. 5D, lower panel). These lysates weresubjected to immunoprecipitation with anti-SHP2 and immu-noblotting with anti-EGFR antibody. As shown in Fig. 5E (toppanel), pervanadate did not affect the trapping of the EGFR bythe T/A-SHP2 or the DM-SHP2 protein even at the higherconcentration used. In fact, it seems to enhance trapping. Asexpected, the WT-SHP2 protein precipitated a very lowamount of the EGFR protein that did not change with PVconcentration. Reblotting with anti-SHP2 antibody showedthat the amount of SHP2 proteins in all lanes was comparable(Fig. 5E, bottom panel). These results indicate that pervana-date does not affect substrate trapping by the T/A. Assumingthat this could be related to low level of pervanadate crossingthe PM, we conducted in vitro substrate trapping in the

Page 8: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

52 R. Merritt et al. / Biochimica et Biophysica Acta 1763 (2006) 45–56

Page 9: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

53R. Merritt et al. / Biochimica et Biophysica Acta 1763 (2006) 45–56

presence of varying concentration of pervanadate. Pervanadatedid not show any effect on substrate trapping even at 10 mMconcentration (data not shown). These results confirm the site-directed mutagenesis studies (Fig. 5B) that Cys459 does notcontribute to substrate trapping by the T/A-SHP2 protein.

3.5. The T/A- and the C/S-T/A-SHP2 proteins are highlyspecific substrate-trapping mutants

The T/A- and the C/S-T/A-SHP2 proteins trap the EGFR asefficiently as the DM. However, it was not clear if they wereselective for Tyr992(P) of the EGFR, which was demonstratedto be an SHP2 target [19]. We thus tested the specificity of theT/A- or the C/S-T/A-SHP2 protein toward Tyr992(P) by affinityprecipitation. Wild type EGFR (WT-EGFR) or Tyr992Phe-EGFR was expressed in NIH-3T3 cells that have undetectablelevel of endogenous EGFR. Cells that were transfected withvector alone were used as negative controls. After serumstarvation for ∼12 h, cells were stimulated with 100 ng/ml EGFfor 10 min. Immunoblot analysis of total cell lysates with anti-EGFR antibody showed that both constructs were expressedcomparably (Fig. 6A, top panel). Reblotting with anti-Tyr(P)antibody further showed that both constructs were Tyrphosphorylated, but there was a slight reduction in theTyr992Phe-EGFR lane reflecting the effect of the mutation(Fig. 6A, middle panel). Note that EGFR has five majorautophosphorylation sites. As a loading control, we blotted thesame membrane with anti-β-actin antibody (Fig. 6A, lowerpanel), which showed the presence of comparable amount ofcellular proteins. These lysates were subjected to affinityprecipitation with the GST fusion of the PTP domain of the T/A,the C/S-T/A or the DM protein. Precipitates were then analyzedby immunoblot with anti-EGFR antibody. As shown in Fig. 6B,these PTP domains affinity precipitated the WT-EGFR, but verylittle or none of the Tyr992Phe-EGFR. Reblotting themembrane with anti-GST antibody showed that there wascomparable amount of the GST fusion protein in each lane.These results suggest that the T/A- and the C/S-T/A-SHP2proteins are highly specific substrate-trapping mutants.

4. Discussion

Unlike other PTPs such as PTP1B and SHP1, SHP2 is aunique PTP in that it positively modulates growth and cellsurvival signals induced by RTKs. However, its mechanism ofaction has been very difficult to uncover primarily because verylittle is known about its target substrates. We have recently

Fig. 5. The role of Cys459 in substrate trapping by the T/A-SHP2 mutant. (A) COconstruct. After incubation for 36 h, cells were serum starved for 12 h and then stimanalyzed by immunoblotting with anti-Tyr(P) (top panel) to examine Tyr phosphoryThe same lysates were immunoprecipitated with anti-SHP2 and blotted with anti-EG1 cells were treated with the indicated amounts of pervanadate (PV) for 30 min. Efwith anti-Tyr(P) antibody (top panel). An anti-actin blot is included as a loading cafter starvation, cells were left untreated or treated with the different concentrationlysates prepared from these cells were analyzed for effect of PV on Tyr phosphorpanel). (E) The same lysates were subjected to immunoprecipitation with anti-SHPtrapping even at 10 mM.

identified the EGFR as a target substrate of SHP2 and haveprovided a mechanism in mediating Ras activation in the EGFRsignalling pathway [42]. More recently, a similar mechanismwas proposed for the Gab1-SHP2 axis in RTK-induced Rasactivation [47], an indication of emerging advances in ourunderstanding of SHP2. In the current report, we show that theconserved residue in the signature motif of SHP2, Thr466, isessential for the catalytic activity of SHP2, and that mutation ofThr466 to Ala provides a new substrate-trapping mutant. Asopposed to the DM- or the C/S-T/A-SHP2 protein, the T/A-SHP2 is a single mutant making it a preferred material forsubstrate identification and structural studies aimed at solvingSHP2 structure in complex with its physiological substrate.

The consensus sequence for the active site signature motif ofsix well-known PTPs is shown in Fig. 1. SHP1, SHP2, R-PTPαand Corkscrew have Thr, while PTP1B and VHR have Serimmediately C-terminal to the invariant Arg residue in thismotif [4,26–30]. However, very little is known about the im-portance of the conserved hydroxyl amino acid in the catalyticprocess. Previous work on VHR has shown that the cor-responding Ser residue mediates the hydrolysis of the cysteinyl-phosphate intermediate [31]. Thus, we hypothesized that Thr466of SHP2 might function a similar way. This possibility wasinvestigated by site-directed mutagenesis, PTPase assays andsubstrate binding studies. The EGFR system was used as amodel for these studies as it is the physiological substrate ofSHP2 [19,42].

SHP2 becomes Tyr phosphorylated upon stimulation of cellswith a variety of growth factors [13,35,36]. One of the uniquecharacteristics of SHP2 is that it auto-regulates its own Tyrphosphorylation [19,37,42]. The anti-Tyr(P) Western blotspresented in Fig. 2A show that the T/A-SHP2 protein wasunable to autodephosphorylate itself suggesting that it is PTPasedefective. These results were further confirmed by PTPaseassays (Fig. 2B and C) that the T/A-SHP2 is in deed a PTPase-dead mutant even when tested at varying pH and time points.Therefore, Thr466 is important for the catalytic activity ofSHP2. Total loss of PTPase activity was unexpected given theprevious report on VHR where mutation of the correspondingSer residue stalled this enzyme in a transition state. Under thoseexperimental conditions, an initial burst of pNP formation wasobserved. In case of SHP2 however, we were unable to detectany activity. These differences might be related to the fact thatSHP2 and VHR are two different proteins with differentphysiological roles.

Mutation of Asp in the WPD loop stalls PTPs in a tran-sition state leading to accumulation of a cysteinyl-phosphate

S-1 cells were transfected with the WT-, T/A-, the C/S-T/A- or the DM-SHP2ulated with EGF for 10 min, 1 h or 2 h. Lysates prepared from these cells werelation or with an anti-SHP2 (bottom panel) antibody to evaluate expression. (B)FR (top panel) or anti-SHP2 (bottom panel) antibody. (C) Serum-starved COS-fect of PV on Tyr phosphorylation of cellular proteins was assessed by blottingontrol (bottom panel). (D) SHP2 proteins were expressed in COS-1 cells, andof PV and then unstimulated or stimulated with EGF for 10 min. Total cell

ylation (top panel). An anti-actin blot is shown as a loading control (bottom2 and immunoblotting with anti-EGFR antibodies. PV did not affect substrate

Page 10: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

Fig. 6. Specificity studies. NIH-3T3 cells were transfected with vector alone, WT-EGFR or Tyr992Phe-EGFR (Y/F). After 36 h of incubation, cells were serum starvedfor 12 h, and then stimulated with EGF for 10 min. (A) Total cell lysates were analyzed for expression of EGFR (top panel) or Tyr phosphorylation (middle panel).Anti-actin blot is included as a loading control. (B) The same lysates were subjected to affinity precipitation with the indicated GST fusion PTPs and analyzed byimmunoblot with anti-EGFR (top panel) or anti-GST (bottom panel).

54 R. Merritt et al. / Biochimica et Biophysica Acta 1763 (2006) 45–56

intermediate. This process requires the presence of thenucleophilic Cys residue that attacks the phosphate moietyof the Tyr(P) leading to formation of a covalent bond[31,44,48,49]. As a result, these mutants become substratetrapping. Mutation of Ser in the signature motif of VHR, aresidue that corresponds to Thr466 of SHP2, also stalls thisenzyme in a transition state [31] although its ability to traptarget substrates was not investigated. In case of SHP2, wehave demonstrated that the T/A-SHP2 traps the EGFR and thethree other unknown phosphotyrosyl proteins (Fig. 3), sameset of proteins trapped by the DM-SHP2 [19]. Therefore, theT/A-SHP2 protein is a substrate trapping mutant. The specificinteraction of the T/A-SHP2 protein with target substratestogether with inhibition studies with OV show that mutationof Thr466 did not cause structural changes that affect substratebinding. As shown in Fig. 4C and D, OV competitivelyinhibited substrate trapping by both the T/A- and DM-SHP2proteins. 1–2 mM OV both in intact cells and in vitroenhanced substrate trapping probably by protecting target Tyr(P) sites, but acted as a competitive inhibitor when used inexcess of this concentration. Thus, substrate trapping solelyusing OV as an inducer of Tyr phosphorylation could becounter productive unless an amount that inhibits binding ispredetermined in each case.

The next logical step was to investigate what role Thr466plays in the catalytic process of SHP2. In the active site of SHP2and other PTPs, the Cys residue acts as a nucleophile, while theArg as a phosphate coordinator. On the other hand, Asp in theWPD loop acts as a proton donor for the leaving phenolategroup and as an acceptor in the hydrolysis of the cysteinylphosphate intermediate. As a result, its mutation to Ala providessubstrate trapping mutants [23,44]. In PTPase assays, thesemutants show initial activity until all enzyme molecules areoccupied. The same is true for Ser131Ala mutant of VHR [31].In case of T/A-SHP2 however, there was total loss of PTPaseactivity indicating that the role of Thr466 might not bemediating the hydrolysis of a transition-state complex sinceno initial para-nitrophenol production was observed in the invitro PTPase reactions (Fig. 2B and C). Clearly, Thr466 is

essential for the catalytic process of SHP2. However, whatspecific role it might play is not clear.

Next, we investigated the importance of Cys459 in substratetrapping by the T/A-SHP2 protein. Differently said, we assessedwhether accumulation of a cysteinyl phosphate intermediatewas the mechanism for the T/A-SHP2 protein to trap substrates.It was reasoned that if nucleophilic attack by Cys459 wereimportant for substrate trapping by the T/A-SHP2, the C/S-T/A-SHP2 should be a weaker trapping mutant for the mutationabolishes the contribution of the T/A leaving only that of theC/S. However, the C/S-T/A-SHP2 was as efficient as the T/A-or the DM-SHP2 in substrate trapping (Fig. 5B) suggestingthat Cys459 may not be important. Since the C/S-SHP2 byitself exhibits substrate-trapping property [20,38,39], ourresults with the C/S-T/A cannot completely rule out theinvolvement of Cys459. Therefore, we conducted substratebinding studies in the presence of varying amounts of PV,which inactivates PTPs by oxidizing the nucleophilic Cysresidue [45,46,50]. In support of the mutagenesis studies, theT/A-SHP2 trapped the EGFR in the presence of PV both inintact cells and outside the cell (Fig. 5E and data not shown).These results show that Cys459 does not contribute tosubstrate trapping by the T/A-SHP2, which in turn suggeststhat Thr466 is not involved in enzyme turnover. It is possiblethat the conserved Ser or Thr residue in the signature motif ofdifferent PTPs may play variable role depending on thecorresponding structural context.

It could be argued that mutation of a conserved residue in theactive site of an enzyme may cause loss of specificity. Ourprevious report demonstrate that the DM-SHP2 protein carryingdouble mutations (Cys459 in the signature motif and Asp425 intheWPD loop) maintains specificity toward target substrates [19].The finding that the T/A-SHP2 protein distinctively recognizesTyr992(P)-EGFR demonstrates retention of substrate specificity.Therefore, the T/A-SHP2 protein is a specific substrate-trappingmutant. Furthermore, the ability of the T/A-SHP2 protein tospecifically bind to target substrates suggests that Thr466 is notinvolved in substrate recognition by the active site. Structuralstudies with PTP1B have shown that the corresponding Ser

Page 11: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

55R. Merritt et al. / Biochimica et Biophysica Acta 1763 (2006) 45–56

residue (Ser222) makes a hydrogen bond with the Sγ atom of thenucleophilic Cys [49]. Similarly, the corresponding Ser residue inthe low molecular weight PTP was proposed to participate instabilizing the nucleophilic Cys [48]. Our findings on SHP2 seemto concur with these studies. Given that the mutation totallyabolished enzyme activity at all pH points tested, and that thismutation did not affect specific interactions with target substrates,we propose that the role of Thr466 in the catalytic process ofSHP2 is to stabilize the sulfhydryl group of Cys459 fornucleophilic attack on the phosphate moiety of the substrate.Obviously, further structural studies are required to verify thispoint. The development of the T/A-SHP2 substrate-trappingmutant with intact nucleophile (Cys459), proton donor andacceptor (Asp425) and the identification of the Tyr992(P) of theEGFR as a substrate of SHP2 [19,42] will provide an excellentstarting material for such studies.

Acknowledgements

We Thank Irene Ischenko for her technical assistance. Thiswork was support in part by West Virginia University internalgrants to YMA, and in part by NIH Public Service GrantsCA28146 to MJH.

References

[1] J. Schlessinger, Cell signaling by receptor tyrosine kinases, Cell 103(2000) 211–225.

[2] J. Schlessinger, M. Mohammadi, B. Margolis, A. Ullrich, Role of SH2-containing proteins in cellular signaling by receptor tyrosine kinases, ColdSpring Harbor Symp. Quant. Biol. 57 (1992) 67–74.

[3] A. Sorkin, K. Helin, C.M. Waters, G. Carpenter, L. Beguinot, Multipleautophosphorylation sites of the epidermal growth factor receptor areessential for receptor kinase activity and internalization. Contrastingsignificance of tyrosine 992 in the native and truncated receptors, J. Biol.Chem. 267 (1992) 8672–8678.

[4] N.K. Tonks, B.G. Neel, Combinatorial control of the specificity of proteintyrosine phosphatases, Curr. Opin. Cell Biol. 13 (2001) 182–195.

[5] A. Alonso, J. Sasin, N. Bottini, I. Friedberg, A. Osterman, A. Godzik, T.Hunter, J. Dixon, T. Mustelin, Protein tyrosine phosphatases in the humangenome, Cell 117 (2004) 699–711.

[6] B.G. Neel, N.K. Tonks, Protein tyrosine phosphatases in signaltransduction, Curr. Opin. Cell Biol. 9 (1997) 193–204.

[7] B.G. Neel, H. Gu, L. Pao, The ‘Shp’ing news: SH2 domain-containingtyrosine phosphatases in cell signaling, Trends Biochem. Sci. 28 (2003)284–293.

[8] G.S. Feng, C.C. Hui, T. Pawson, SH2-containing phosphotyrosinephosphatase as a target of protein-tyrosine kinases, Science 259 (1993)1607–1611.

[9] G.S. Feng, T. Pawson, Phosphotyrosine phosphatases with SH2 domains:regulators of signal transduction, Trends Genet. 10 (1994) 54–58.

[10] D. D'Ambrosio, K.L. Hippen, S.A. Minskoff, I. Mellman, G. Pani, K.A.Siminovitch, J.C. Cambier, Recruitment and activation of PTP1C innegative regulation of antigen receptor signaling by Fc gamma RIIB1,Science 268 (1995) 293–297.

[11] S. Krautwald, D. Buscher, V. Kummer, S. Buder, M. Baccarini,Involvement of the protein tyrosine phosphatase SHP-1 in Ras-mediatedactivation of the mitogen-activated protein kinase pathway, Mol. Cell.Biol. 16 (1996) 5955–5963.

[12] S.M. Keyse, Protein phosphatases and the regulation of mitogen-activatedprotein kinase signalling, Curr. Opin. Cell Biol. 12 (2000) 186–192.

[13] R.J. Lechleider, R.M. Freeman Jr., B.G. Neel, Tyrosyl phosphorylation andgrowth factor receptor association of the human corkscrew homologue,SH-PTP2, J. Biol. Chem. 268 (1993) 13434–13438.

[14] J.D. Allard, H.C. Chang, R. Herbst, H. McNeill, M.A. Simon, The SH2-containing tyrosine phosphatase corkscrew is required during signaling bysevenless, Ras1 and Raf, Development 122 (1996) 1137–1146.

[15] V. Cleghon, P. Feldmann, C. Ghiglione, T.D. Copeland, N. Perrimon, D.A.Hughes, D.K. Morrison, Opposing actions of CSWand RasGAP modulatethe strength of Torso RTK signaling in the Drosophila terminal pathway,Mol. Cell 2 (1998) 719–727.

[16] A.M. Bennett, S.F. Hausdorff, A.M. O'Reilly, R.M. Freeman, B.G. Neel,Multiple requirements for SHPTP2 in epidermal growth factor-mediatedcell cycle progression, Mol. Cell. Biol. 16 (1996) 1189–1202.

[17] T.M. Saxton, M. Henkemeyer, S. Gasca, R. Shen, D.J. Rossi, F. Shalaby,G.S. Feng, T. Pawson, Abnormal mesoderm patterning in mouse embryosmutant for the SH2 tyrosine phosphatase Shp-2, EMBO J. 16 (1997)2352–2364.

[18] T.M. Saxton, B.G. Ciruna, D. Holmyard, S. Kulkarni, K. Harpal, J.Rossant, T. Pawson, The SH2 tyrosine phosphatase shp2 is required formammalian limb development, Nat. Genet. 24 (2000) 420–423.

[19] Y.M. Agazie, M.J. Hayman, Development of an efficient “substrate-trapping” mutant of Src homology phosphotyrosine phosphatase 2 andidentification of the epidermal growth factor receptor, Gab1, and threeother proteins as target substrates, J. Biol. Chem. 278 (2003)13952–13958.

[20] Z.Y. Zhang, Y. Wang, L. Wu, E.B. Fauman, J.A. Stuckey, H.L. Schubert,M.A. Saper, J.E. Dixon, The Cys(X)5Arg catalytic motif in phosphoesterhydrolysis, Biochemistry 33 (1994) 15266–15270.

[21] J. Yuvaniyama, J.M. Denu, J.E. Dixon, M.A. Saper, Crystal structure of thedual specificity protein phosphatase VHR, Science 272 (1996)1328–1331.

[22] L. Xie, Y.L. Zhang, Z.Y. Zhang, Design and characterization of animproved protein tyrosine phosphatase substrate-trapping mutant, Bio-chemistry 41 (2002) 4032–4039.

[23] A.J. Flint, T. Tiganis, D. Barford, N.K. Tonks, Development of and quot;substrate-trapping and quot; mutants to identify physiological substrates ofprotein tyrosine phosphatases, Proc. Natl. Acad. Sci. U. S. A. 94 (1997)1680–1685.

[24] T. Tiganis, A.J. Flint, S.A. Adam, N.K. Tonks, Association of the T-cellprotein tyrosine phosphatase with nuclear import factor p97, J. Biol. Chem.272 (1997) 21548–21557.

[25] H. Wheadon, N.R. Paling, M.J. Welham, Molecular interactions of SHP1and SHP2 in IL-3-signalling, Cell. Signal. 14 (2002) 219–229.

[26] L.A. Perkins, I. Larsen, N. Perrimon, corkscrew encodes a putativeprotein tyrosine phosphatase that functions to transduce the terminalsignal from the receptor tyrosine kinase torso, Cell 70 (1992)225–236.

[27] T. Yi, J.L. Clevel, J.N. Ihle, Identification of novel protein tyrosinephosphatases of hematopoietic cells by polymerase chain reactionamplification, Blood 78 (1991) 2222–2228.

[28] P. Hof, S. Pluskey, S. Dhe-Paganon, M.J. Eck, S.E. Shoelson,Crystal structure of the tyrosine phosphatase SHP-2, Cell 92 (1998)441–450.

[29] A.M. Bilwes, J. den Hertog, T. Hunter, J.P. Noel, Structural basis forinhibition of receptor protein-tyrosine phosphatase-alpha by dimerization,Nature 382 (1996) 555–559.

[30] R.M. Freeman Jr., J. Plutzky, B.G. Neel, Identification of a human srchomology 2-containing protein-tyrosine-phosphatase: a putative homologof Drosophila corkscrew, Proc. Natl. Acad. Sci. U. S. A. 89 (1992)11239–11243.

[31] J.M. Denu, J.E. Dixon, A catalytic mechanism for the dual-specificphosphatases, Proc. Natl. Acad. Sci. U. S. A. 92 (1995) 5910–5914.

[32] C.Y. Park, M.J. Hayman, The tyrosines in the bidentate motif of the env-sea oncoprotein are essential for cell transformation and are binding sitesfor Grb2 and the tyrosine phosphatase SHP-2, J. Biol. Chem. 274 (1999)7583–7590.

[33] Y.M. Agazie, N. Movilla, I. Ischenko, M.J. Hayman, The phosphotyrosinephosphatase SHP2 is a critical mediator of transformation induced by the

Page 12: Mutation of Thr466 in SHP2 abolishes its phosphatase activity, but provides a new substrate-trapping mutant

56 R. Merritt et al. / Biochimica et Biophysica Acta 1763 (2006) 45–56

oncogenic fibroblast growth factor receptor 3, Oncogene 22 (2003)6909–6918.

[34] S.J. Taylor, D. Shalloway, Cell cycle-dependent activation of Ras, Curr.Biol. 6 (1996) 1621–1627.

[35] A.M. Bennett, T.L. Tang, S. Sugimoto, C.T. Walsh, B.G. Neel, Protein-tyrosine-phosphatase SHPTP2 couples platelet-derived growth factorreceptor beta to Ras, Proc. Natl. Acad. Sci. U. S. A. 91 (1994) 7335–7339.

[36] W. Lu, D. Gong, D. Bar-Sagi, P.A. Cole, Site-specific incorporation of aphosphotyrosine mimetic reveals a role for tyrosine phosphorylation ofSHP-2 in cell signaling, Mol. Cell 8 (2001) 759–769.

[37] M. Stein-Gerlach, A. Kharitonenkov, W. Vogel, S. Ali, A. Ullrich, Protein-tyrosine phosphatase 1D modulates its own state of tyrosine phosphory-lation, J. Biol. Chem. 270 (1995) 24635–24637.

[38] G. Zhou, J.M. Denu, L. Wu, J.E. Dixon, The catalytic role of Cys124 in thedual specificity phosphatase VHR, J. Biol. Chem. 269 (1994)28084–28090.

[39] Z.Y. Zhang, Y. Wang, J.E. Dixon, Dissecting the catalytic mechanism ofprotein-tyrosine phosphatases, Proc. Natl. Acad. Sci. U. S. A. 91 (1994)1624–1627.

[40] W.L. Chen, D.L. Harris, N.C. Joyce, Effects of SOV-induced phosphataseinhibition and expression of protein tyrosine phosphatases in rat cornealendothelial cells, Exp. Eye Res. 81 (2005) 570–580.

[41] M.C. Zatelli, D. Piccin, F. Tagliati, A. Bottoni, A. Luchin, E.C. degliUberti, SRC homology-2-containing protein tyrosine phosphatase-1restrains cell proliferation in human medullary thyroid carcinoma,Endocrinology 146 (2005) 2692–2698.

[42] Y.M. Agazie, M.J. Hayman, Molecular mechanism for a role of SHP2 in

epidermal growth factor receptor signaling, Mol. Cell. Biol. 23 (2003)7875–7886.

[43] A.J. Garton, A.J. Flint, N.K. Tonks, Identification of p130(cas) as asubstrate for the cytosolic protein tyrosine phosphatase PTP-PEST, Mol.Cell. Biol. 16 (1996) 6408–6418.

[44] J.M. Denu, G. Zhou, Y. Guo, J.E. Dixon, The catalytic role of aspartic acid-92 in a human dual-specific protein-tyrosine-phosphatase, Biochemistry34 (1995) 3396–3403.

[45] H. Deng, R. Callender, Z. Huang, Z.Y. Zhang, Is the PTPase–vanadatecomplex a true transition state analogue? Biochemistry 41 (2002)5865–5872.

[46] L. Zhu, A. Zilbering, X. Wu, K. Mahadev, J.I. Joseph, S. Jabbour, W.Deeb, B.J. Goldstein, Use of an anaerobic environment to preserve theendogenous activity of protein-tyrosine phosphatases isolated from intactcells, FASEB J. 15 (2001) 1637–1639.

[47] A. Montagner, A. Yart, M. Dance, B. Perret, J.P. Salles, P. Raynal, A novelrole for Gab1 and SHP2 in epidermal growth factor-induced Rasactivation, J. Biol. Chem. 280 (2005) 5350–5360.

[48] K. Kolmodin, J. Aqvist, The catalytic mechanism of protein tyrosinephosphatases revisited, FEBS Lett. 498 (2001) 208–213.

[49] A.D. Pannifer, A.J. Flint, N.K. Tonks, D. Barford, Visualization of thecysteinyl-phosphate intermediate of a protein-tyrosine phosphatase byX-ray crystallography, J. Biol. Chem. 273 (1998) 10454–10462.

[50] G. Huyer, S. Liu, J. Kelly, J. Moffat, P. Payette, B. Kennedy, G. Tsaprailis,M.J. Gresser, C. Ramachandran, Mechanism of inhibition of protein-tyrosine phosphatases by vanadate and pervanadate, J. Biol. Chem. 272(1997) 843–851.