pYtags enable spatiotemporal measurements of receptor tyrosine kinase signaling in living cells

  1. Payam E Farahani
  2. Xiaoyu Yang
  3. Emily V Mesev
  4. Kaylan A Fomby
  5. Ellen H Brumbaugh-Reed
  6. Caleb J Bashor  Is a corresponding author
  7. Celeste M Nelson  Is a corresponding author
  8. Jared E Toettcher  Is a corresponding author
  1. Department of Chemical & Biological Engineering, Princeton University, United States
  2. Department of Bioengineering, Rice University, United States
  3. Program in Systems, Synthetic, and Physical Biology, Rice University, United States
  4. Department of Molecular Biology, Princeton University, United States
  5. IRCC International Research Collaboration Center, National Institutes of Natural Sciences, Japan
  6. Department of Biosciences, Rice University, United States
6 figures, 3 tables and 1 additional file

Figures

Figure 1 with 5 supplements
pYtags: a biosensing strategy to monitor receptor tyrosine kinase (RTK) activity in living cells.

(A) The T-cell receptor complex contains six immunoreceptor tyrosine-based activation motifs (ITAMs) from CD3 chains that, when phosphorylated, bind to the tSH2 domain of ZAP70 (ZtSH2). (B) Three repeats of CD3 ITAMs were appended to the C-terminus of epidermal growth factor receptor (EGFR) and clearance of ZtSH2 from the cytosol was assessed. (C) Timelapse images of NIH3T3 cells expressing EGFR pYtag (CD3ε variant), treated first with EGF (100 ng/mL) and then with gefitinib (10 µM). Scale bar, 20 µm. (D) Mean clearance of cytosolic ZtSH2 in cells co-expressing iRFP-ZtSH2 and EGFR C-terminally labeled with one of six CD3 ITAMs. EGF (100 ng/mL) and gefitinib (10 µM) were sequentially added at times denoted by arrows. n = 2 independent experiments. (E) Clearance of cytosolic ZtSH2 10 min post-EGF treatment and 40 min post-gefitinib treatment from (D). Lines denote mean values, boxes denote 25–75th percentiles, and whiskers denote minima and maxima. n 14 cells from two independent experiments. n.s., not significant, ***p<0.001 by Kolmogorov–Smirnov test with cells expressing no additional EGFR 10 min post-EGF. (F) Immunoblots of NIH3T3 cells expressing either WT EGFR or EGFR pYtag treated with EGF (100 ng/mL). (G) Mean ± SEM levels of EGFR, Akt, and Erk phosphorylation from (F). n = 3 independent experiments. (H) The EGFR pYtag was tested in SYF cells to determine whether SFKs are required for ITAM phosphorylation. (I) Representative images of NIH3T3 and SYF cells expressing EGFR pYtag, treated with EGF (100 ng/mL). Scale bars, 40 µm. (J) Mean clearance of cytosolic ZtSH2 in SYF and NIH3T3 cells 10 min after treatment with EGF. For each condition, n > 20 cells from three independent experiments. See also Figure 1—video 1.

Figure 1—figure supplement 1
Grb2 fails to discriminate between immunoreceptor tyrosine-based activation motif (ITAM)-labeled and unlabeled RTKs.

(A) ZtSH2- and Grb2-based reporters of receptor tyrosine kinase (RTK) signaling were co-expressed in cells expressing either ITAM-labeled or WT epidermal growth factor receptor (EGFR). (B) NIH3T3 cells before and 5 min after treatment with EGF (100 ng/mL). Scale bars, 20 µm. (C) Mean ± SD clearance of ZtSH2 and Grb2 reporters from the cytosol following treatment with EGF (100 ng/mL). n = 3 independent experiments. (D) Clearance of reporters from the cytosol 5 min after treatment with EGF in (C). Lines denote mean values, boxes denote 25–75th percentiles, and whiskers denote minima and maxima. For each condition, n > 16 cells from three independent experiments. (E) ZtSH2 reports the signaling of ITAM-labeled RTKs, while Grb2 fails to discriminate between RTKs displaying or lacking ITAMs.

Figure 1—figure supplement 2
Effects of the expression levels of pYtag components in NIH3T3 cells.

Images of EGFR-pYtag expressing NIH3T3 cells expressing various levels of (A) EGFR-CD3ε-FusionRed or (B) iRFP-ZtSH2, treated with EGF (20 ng/mL). Scale bars, 20 µm. (C) Maximum % clearance of ZtSH2 from the cytosol in EGFR-pYtag expressing NIH3T3 cells treated with EGF (20 ng/mL). (D) Maximum % clearance of ZtSH2 from the cytosol as a function of the ratio of EGFR-CD3ε-FusionRed to iRFP-ZtSH2 (EGFR:ZtSH2 ratio). (E, F) Heatmaps of single-cell trajectories for all cells quantified in (C) and sorted in order of increasing EGFR:ZtSH2 ratio. (E) shows the % clearance of ZtSH2 from the cytosol; (F) shows min–max normalized responses to compare activation kinetics. Panels (C–E): points denote individual cells. n = 86 cells from three independent experiments.

Figure 1—figure supplement 3
Effects of the expression levels of pYtag components in HEK293T cells.

Images of EGFR-pYtag expressing HEK293T cells expressing variable levels of (A) EGFR-CD3ε-mNeonGreen or (B) mScarlet-ZtSH2, treated with EGF (100 ng/mL). Scale bars, 20 µm. (C) Maximum % clearance of ZtSH2 from the cytosol in EGFR-pYtag expressing HEK293T cells treated with EGF (100 ng/mL). (D) Maximum % clearance of ZtSH2 from the cytosol as a function of the ratio of EGFR-CD3ε-mNeonGreen to mScarlet-ZtSH2 (EGFR:ZtSH2 ratio). (E, F) Heatmaps of single-cell trajectories for all cells quantified in (C) and sorted in order of increasing EGFR:ZtSH2 ratio. (E) shows the % clearance of ZtSH2 from the cytosol; (F) shows min–max normalized responses to compare activation kinetics. Panels (C, D): points denote individual cells. n = 84 cells from three independent experiments.

Figure 1—figure supplement 4
Mathematical modeling of the effects of pYtag component expression levels on biosensor readout.

(A) Heatmap of maximum % clearance of ZtSH2 in response to 20 ng/mL EGF. For each cell, rows and columns denote levels of EGFR-CD3ε and ZtSH2, respectively. (B) Each cell from (A) was plotted as a function of the ratio of EGFR to ZtSH2 (EGFR:ZtSH2 ratio). Right plot presents zoomed-in results for EGFR:ZtSH2 ratios between 0.5 and 2.

Figure 1—video 1
Timelapse of iRFP-ZtSH2 in NIH3T3 cells co-expressing iRFP-ZtSH2 and EGFR-CD3ε-FusionRed.

Cells were first treated with EGF (100 ng/mL) then treated with gefitinib (10 µM) at the times denoted in the video. Related to Figure 1C.

Figure 2 with 2 supplements
Monitoring epidermal growth factor receptor (EGFR) signaling at subcellular and multicellular length scales.

(A) Images of EGFR pYtag-expressing NIH3T3 cells treated with EGF (20 ng/mL). Scale bar, 20 µm. (B) Mean ± SD clearance of cytosolic ZtSH2 from (A). n = 3 independent experiments. (C) EGFR pYtag-expressing NIH3T3 cells treated with EGF (20 ng/mL) were monitored for internalized ZtSH2-positive vesicles, and then treated with gefitinib (10 µM). Scale bar, 20 µm. (D) Timelapse images from the region denoted by the blue dashed border from (C). Scale bar, 10 µm. (E) MCF10A human mammary epithelial cells cultured on soft substrata form round, multilayered clusters. EGFR pYtag and ErkKTR were used to spatiotemporally monitor both EGFR and Erk responses after stimulation with EGF. (F) Images of MCF10A cells cultured on soft substrata and treated with EGF (100 ng/mL). Scale bar, 25 µm. (G) Apical and lateral enrichment of ZtSH2 was quantified by line scans denoted by magenta and orange vectors, respectively. Scale bar, 10 µm. (H) Heatmaps of EGFR pYtag and ErkKTR responses from (F). Rows denote individual cells. For each cell, signaling responses of each biosensor were normalized to their respective minima and maxima. (I) Time to half maximal response for EGFR pYtag and ErkKTR for cells from (H). n = 19 (periphery) and n = 11 (interior) cells from three biological replicates. (J) Time to half maximal response for EGFR pYtag and ErkKTR. Responses of individual cells are denoted by points and connected by lines. n > 30 cells from three biological replicates. ***p<0.001 by Kolmogorov–Smirnov test. See also Figure 2—video 1.

Figure 2—figure supplement 1
Analysis of epidermal growth factor receptor (EGFR) and ZtSH2 internalization.

(A) Images of control (EGFR-FusionRed) and EGFR pYtag (EGFR-CD3ε-FusionRed; iRFP-ZtSH2) NIH3T3 cells immunostained for EEA1 after treatment with EGF (100 ng/mL) for varying durations. Scale bars, 20 µm. (B) Intensity of EGFR-CD3ε-FusionRed at the cell membrane from (A). For each condition, n > 70 cells from three independent experiments. (C) Volume of pixels doubly positive for EGFR-CD3ε-FusionRed and EEA1 in individual cells. For each condition, n > 36 cells from three independent experiments. (D) Image of EGFR pYtag-expressing NIH3T3 cell treated with EGF (100 ng/mL) for 30 min and immunostained for ZtSH2 and EEA1. Scale bars, 20 µm (top) and 5 µm (bottom). (E) Fluorescence measurements along the red vector in (D). For each protein, intensities were normalized to the minimum and maximum values measured. (F) Image of EGFR pYtag-expressing HEK293T cell treated with EGF (100 ng/mL) for 5 min and immunostained for EEA1. Scale bars, 10 µm (top) and 3 µm (bottom). (G) Fluorescence measurements along the red vector in (F). For each protein, intensities were normalized to the minimum and maximum values measured.

Figure 2—video 1
Maximum intensity projection timelapse images of MCF10A cells co-expressing iRFP-ZtSH2 (left panel), EGFR-CD3ε-FusionRed (middle panel), and ErkKTR-BFP (right panel), cultured on soft substrata and treated with EGF (100 ng/mL).

Related to Figure 2F.

Figure 3 with 2 supplements
Epidermal growth factor receptor (EGFR) pYtag reveals dose- and ligand-dependent signaling dynamics.

(A) Mean ± SD responses of EGFR pYtag-expressing NIH3T3 cells to varying doses of EGF, epiregulin (EREG), and epigen (EPGN). The same 0 ng/mL control was used for each ligand. Data were collected from 475 cells across four independent experiments with each dose tested at least twice. (B) Dose–response profiles from (A) were analyzed using a mathematical model of EGFR pYtag. (C) Simulations of EGFR pYtag responses to ligand of varying doses for different values of β and γ. (D) GBM-associated mutants of EGFR that form strong EREG-bound dimers were predicted to exhibit stronger pYtag responses to 20 ng/mL EREG compared to WT EGFR. (E) Mean ± SD pYtag response of WT and GBM-associated mutant EGFRs in NIH3T3 cells after EREG treatment (20 ng/mL). n = 3 independent experiments.

Figure 3—figure supplement 1
ZtSH2 and epidermal growth factor receptor (EGFR) localization in response to different ligands.

Representative images of EGFR pYtag-expressing NIH3T3 cells treated with different concentrations of EGF, EREG, or EPGN. Scale bars, 20 µm.

Figure 3—figure supplement 2
Ligand-free dimers in mathematical model recapitulate biphasic signaling response of epidermal growth factor receptor (EGFR).

(A) EGFR pYtag response to EGF (20 ng/mL) shown in Figure 2B. (B) The effect of ligand-free dimers on EGFR pYtag responses was simulated by tuning the dissociation rate of ligand-free dimers k6. (C) Percentage of receptors existing as ligand-free dimers before ligand stimulation as a function of k6. Color of data points corresponds to color of curves in (D). (D) Simulated EGFR pYtag responses to EGF for varying values of k6.

Figure 4 with 1 supplement
Monitoring distinct receptor tyrosine kinases (RTKs) in heterodimeric complexes.

(A) In order to signal, the ligandless ErbB2 must heterodimerize with a ligand-binding member of the ErbB family. The pYtag strategy enables measurements of ErbB2’s activity despite the co-activation of epidermal growth factor receptor (EGFR). (B) Representative images of NIH3T3 cells treated with EGF (100 ng/mL). Scale bar, 20 µm. (C) Mean ± SD clearance of cytosolic ZtSH2 after treatment with EGF (100 ng/mL). n = 3 independent experiments.

Figure 4—figure supplement 1
pYtag biosensors of additional receptor tyrosine kinases (RTKs).

(A) Images of NIH3T3 cells co-expressing iRFP-ZtSH2 with either FGFR1-FusionRed or FGFR1-CD3ε-FusionRed, treated with FGF4 (100 ng/mL). Scale bars, 20 µm. (B) Mean ± SD clearance of cytosolic ZtSH2 in cells expressing FGFR1-FusionRed (ctrl) or FGFR1-CD3ε-FusionRed (CD3ε) from (A). n = 3 independent experiments. (C) Clearance of ZtSH2 from the cytosol 10 min after treatment with FGF4 in (B). (D) Images of NIH3T3 cells co-expressing iRFP-ZtSH2 with either PDGFRβ-FusionRed or PDGFRβ-CD3ε-FusionRed, treated with PDGF-BB (100 ng/mL). Scale bars, 20 µm. (E) Mean ± SD clearance of cytosolic ZtSH2 in cells expressing PDGFRβ-FusionRed (ctrl) or PDGFRβ-CD3ε-FusionRed (CD3ε) from (D). n = 3 independent experiments. (F) Clearance of ZtSH2 from the cytosol 10 min after treatment with PDGF-BB in (E). (G) Images of NIH3T3 cells co-expressing iRFP-ZtSH2 with either VEGFR3-FusionRed or VEGFR3-CD3ε-FusionRed, treated with VEGF-C (500 ng/mL). Scale bars, 20 µm. (H) Mean ± SD clearance of cytosolic ZtSH2 in cells expressing VEGFR3-FusionRed (ctrl) or VEGFR3-CD3ε-FusionRed (CD3ε) from (G). n = 3 independent experiments. (I) Clearance of ZtSH2 from the cytosol 10 min after treatment with VEGF-C in (H). Lines denote mean values, boxes denote 25–75th percentiles, and whiskers denote minima and maxima. For each condition, n > 38 cells from three independent experiments. ***p<0.001 by Kolmogorov–Smirnov test.

Figure 5 with 3 supplements
Orthogonal pYtags enable multiplexed receptor tyrosine kinase (RTK) biosensing.

(A) To assess the performance of the VISH2/SLP76 system as a pYtag-based biosensor, VISH2 and ZtSH2 reporters were co-expressed in NIH3T3 cells along with either SLP76- or CD3ε-labeled epidermal growth factor receptor (EGFR). (B) NIH3T3 cells co-expressing VISH2 and ZtSH2 reporters before and 3 min after treatment with EGF (100 ng/mL). Scale bars, 20 µm. (C) Mean ± SD clearances of VISH2 and ZtSH2 from the cytosol, expressed with either SLP76- or CD3ε-labeled EGFR and stimulated with EGF (100 ng/mL). n = 3 independent experiments. (D) Response of VISH2 and ZtSH2 reporters 10 min after EGF treatment in (C). n > 30 cells from three independent experiments. (E) Orthogonal pYtags can be multiplexed to monitor the activity of multiple RTKs in the same cell. (F) Images of cells expressing orthogonal reporters for EGFR and ErbB2, treated with EGF (100 ng/mL). Scale bar, 20 µm. (G) Mean ± SD trajectories for EGFR and ErbB2 activity using multiplexed pYtags. For each reporter, the mean response was normalized to its minimum and maximum measured values. n = 3 independent experiments. (H) Time to half maximal response for individual cells from (G). Lines denote mean values, boxes denote 25–75th percentiles, and whiskers denote minima and maxima. n > 30 cells from three independent experiments. ***p<0.001 by Kolmogorov–Smirnov test. See also Figure 5—video 1 and Figure 5—video 2.

Figure 5—figure supplement 1
Comparison of epidermal growth factor receptor (EGFR) and ErbB2 responses across experiments.

(A) Mean responses of EGFR and ErbB2 to EGF (100 ng/mL) were collected from experiments with or without multiplexed biosensors (see legend at bottom right of figure) and (B) normalized to their minimum and maximum responses for comparison. (C) Time to half maximal response from (B). Lines denote mean values, boxes denote 25–75th percentiles, and whiskers denote minima and maxima. For each condition, n ≥ 30 cells from three independent experiments.

Figure 5—video 1
Timelapse of NIH3T3 cells co-expressing VISH2 and ZtSH2 reporters, and either SLP76- or CD3ε-labeled epidermal growth factor receptor (EGFR), treated with EGF (100 ng/mL).

Related to Figure 5B.

Figure 5—video 2
Timelapse of NIH3T3 cells co-expressing pYtag biosensors for epidermal growth factor receptor (EGFR) and ErbB2, treated with EGF (100 ng/mL).

Related to Figure 5F.

Figure 6 with 1 supplement
pYtags can be used to monitor the activity of endogenous receptor tyrosine kinases (RTKs).

(A) Schematic of EGFR locus containing the C-terminus of epidermal growth factor receptor (EGFR), where CRISPR/Cas9 was used to label the receptor with CD3ε-mNeonGreen via homology-directed repair. (B) PCR of genomic DNA from parental or knock-in HEK293T cells. Validation primers targeting homology regions upstream and downstream of the CD3ε-mNeonGreen insert are labeled by black arrows in (A). (C) Immunoblots of EGFR in parental or knock-in HEK293T cells. (D) Images of parental or knock-in HEK293T cells treated with EGF (100 ng/mL). mScarlet-ZtSH2 images show averages of two successive frames to decrease background noise; full raw movie is included as Figure 6—video 1. Scale bar, 10 µm. (E) Mean ± SD clearance of ZtSH2 from the cytosol following treatment with EGF (100 ng/mL). parental HEK293T, n = 3 independent experiments; knock-in HEK293T, n = 4 independent experiments. (F) Clearance of ZtSH2 from the cytosol 1 min after treatment with EGF in (E). Lines denote mean values, boxes denote 25–75th percentiles, and whiskers denote minima and maxima. parental HEK293T, n = 23 cells from three independent experiments; knock-in HEK293T, n = 46 cells from four independent experiments. ***p<0.001 by Kolmogorov–Smirnov test. See also Figure 6—video 1.

Figure 6—video 1
Timelapse of HEK293T cells expressing mScarlet-ZtSH2 and an endogenously labeled EGFR-CD3ε-mNeonGreen, treated with EGF (100 ng/mL).

Left panel shows mScarlet fluorescence; right panel shows mNeonGreen fluorescence at the indicate time points. Scale bar, 20 μm. Related to Figure 6D.

Tables

Appendix 1—key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
Cell line (human)MCF10A-5EJanes et al., 2010RRID:CVCL_0598
Cell line (human)HEK293T LXClonTech LaboratoriesCat # 632180
Cell line (mouse)NIH3T3ATCCCat # CRL-1658
Cell line (mouse)SYF mouse embryonic fibroblasts (MEFs)ATCCCat # CRL-2459
Cell line (Escherichia coli)Stellar chemically competent cellsClonTech LaboratoriesCat # 636763
Recombinant DNA reagentpCMV-dR8.91 lentivirus packaging plasmidGift from Prof. Didier Trono, EPFLAddgene # 12263
Recombinant DNA reagentpMD2.G lenti helper plasmidGift from Prof. Didier Trono, EPFLAddgene # 12259
Recombinant DNA reagentpHR EGFR-FusionRedYang et al., 2021Addgene # 179263
Recombinant DNA reagentpHR EGFR-CD3ζ1-FusionRedThis paperN/AEGFR-ITAM construct
Recombinant DNA reagentpHR EGFR-CD3ζ2-FusionRedThis paperN/AEGFR-ITAM construct
Recombinant DNA reagentpHR EGFR-CD3ζ3-FusionRedThis paperN/AEGFR-ITAM construct
Recombinant DNA reagentpHR EGFR-CD3γ-FusionRedThis paperN/AEGFR-ITAM construct
Recombinant DNA reagentpHR EGFR-CD3δ-FusionRedThis paperN/AEGFR-ITAM construct
Recombinant DNA reagentpHR EGFR-CD3ε-FusionRedThis paperAddgene # 188626EGFR-ITAM construct
Recombinant DNA reagentpHR EGFR- CD3ε-mNeonGreenThis paperN/AEGFR-ITAM construct
Recombinant DNA reagentpHR iRFP-ZtSH2This paperAddgene # 188627ZtSH2 biosensor
Recombinant DNA reagentpHR mScarlet-ZtSH2This paperN/AZtSH2 biosensor
Recombinant DNA reagentpHR EGFR(R84K)-CD3ε-FusionRedThis paperN/AGBM-mutant EGFR construct (Figure 3)
Recombinant DNA reagentpHR EGFR(A265V)-CD3ε-FusionRedThis paperN/AGBM-mutant EGFR construct
(Figure 3)
Recombinant DNA reagentpHR ErbB2-FusionRedThis paperN/AITAM-less ErbB2 construct
(Figure 4)
Recombinant DNA reagentpHR ErbB2- CD3ε-FusionRedThis paperAddgene # 188628ITAM-tagged ErbB2 (Figure 4)
Recombinant DNA reagentpHR EGFR-CitrineThis paperN/AFluorescent EGFR construct
(Figure 4)
Recombinant DNA reagentpHR Clover-VISH2This paperAddgene # 188629tSH2 biosensor
(Figure 5)
Recombinant DNA reagentpHR EGFR-CD3ε-TagBFPThis paperN/AITAM-tagged EGFR (Figure 5)
Recombinant DNA reagentpHR EGFR-SLP76-TagBFPThis paperAddgene # 188630ITAM-tagged EGFR (Figure 5)
Recombinant DNA reagentpHR Grb2-TagBFPThis paperAddgene # 188631Grb2-based biosensor
(Figure 1)
Recombinant DNA reagentpHR FGFR1-CD3ε-FusionRedThis paperAddgene # 188632ITAM-tagged FGFR1
(Figure 4)
Recombinant DNA reagentpHR FGFR1-FusionRedThis paperN/AITAM-less FGFR1
(Figure 4)
Recombinant DNA reagentpHR PDGFRβ-CD3ε-FusionRedThis paperN/AITAM-tagged PDGFR
(Figure 4)
Recombinant DNA reagentpHR PDGFRβ-FusionRedThis paperN/AITAM-less PDGFR
(Figure 4)
Recombinant DNA reagentpHR VEGFR3-CD3ε-FusionRedThis paperN/AITAM-tagged VEGFR
(Figure 4)
Recombinant DNA reagentpHR VEGFR3-FusionRedThis paperN/AITAM-less VEGFR
(Figure 4)
Recombinant DNA reagentpHR ErkKTR-TagBFP9N/A
Recombinant DNA reagentpX330 EGFR-sgRNAThis paper, using a plasmid from Feng Zhang, MITAddgene # 188633EGFR-targeting gRNA (Figure 6)
Recombinant DNA reagentpUC19 EGFRup-CD3ε-mNeonGreen-EGFRdownThis paperAddgene # 188634CRISPR plasmid for EGFR modification (Figure 6)
Sequence-based reagentPEF122 forward primerThis paper5′- TTCTTTTGCAGCAACAGCAAGAGGGCCCTCCC-3′Used to verify CRISPR tagging; see ‘Methods’
Sequence-based reagentPEF123 reverse primerThis paper5’- TCCGTTTCTTCTTTGCCCAGGAAGGGACAGAGTGGCTTATCC-3’Used to verify CRISPR tagging; see ‘Methods’
AntibodyAnti-EGFR antibody (rabbit monoclonal)Cell Signaling TechnologyCat # 4267Used at 1:1000 for western blotting
AntibodyAnti-pEGFR antibody (rabbit monoclonal)Cell Signaling TechnologyCat # 3777Used at 1:1000 for western blotting
AntibodyAnti-β-actin antibody (mouse monoclonal)Cell Signaling TechnologyCat # 3700Used at 1:1000 for western blotting
AntibodyAnti-pAkt antibody (rabbit polyclonal)Cell Signaling TechnologyCat # 9271Used at 1:1000 for western blotting
AntibodyAnti-ppErk antibody (rabbit polyclonal)Cell Signaling TechnologyCat # 9101Used at 1:1000 for western blotting
AntibodyAnti-EEA1 antibody (mouse monoclonal)Cell Signaling TechnologyCat # 48453Used at 1:100 for immunostaining
AntibodyAnti-ZAP70 antibody (rabbit monoclonal)Cell Signaling TechnologyCat # 3165Used at 1:1000 for western blotting
AntibodyGoat anti-Mouse IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 488
(goat polyclonal)
InvitrogenCat # A-11001Used at 1:500 for immunostaining
AntibodyGoat anti-Rabbit IgG (Heavy chain), Superclonal Recombinant Secondary Antibody, Alexa Fluor 647
(goat polyclonal)
InvitrogenCat # A27040Used at 1:500 for immunostaining
AntibodyGoat anti-Mouse IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 647
(goat polyclonal)
InvitrogenCat # A-21236Used at 1:500 for immunostaining
AntibodyIRDye 680RD Goat anti-Mouse IgG antibody
(goat polyclonal)
LI-CORCat # 926-68070Used at 1:10,000 for western blotting
AntibodyIRDye 800CW Goat anti-Rabbit IgG antibody
(goat polyclonal)
LI-CORCat # 926-32211Used at 1:10,000 for western blotting
Peptide, recombinant proteinBovine serum albuminSigma-AldrichCat # 12659
Peptide, recombinant proteinFibronectinCorningCat # CB-40008ACell adhesion coating
Peptide, recombinant proteinClonAmp HiFi PCR polymeraseClonTech LaboratoriesCat # 639298Polymerase
Peptide, recombinant proteinInsulinSigma-AldrichCat # I6634
Peptide, recombinant proteinCholera toxinSigma-AldrichCat # C8052
Peptide, recombinant proteinL-glutamineGibcoCat # 25030-081
Peptide, recombinant proteinEGFR&D SystemsCat # 236-EG-200
Peptide, recombinant proteinEpiregulinR&D SystemsCat # 1195-EP-025
Peptide, recombinant proteinEpigenR&D SystemsCat # 6629-EP-025
Peptide, recombinant proteinFGF4R&D SystemsCat # 235-F4-025
Peptide, recombinant proteinPDGF-BBMillipore SigmaCat # P3201
Peptide, recombinant proteinVEGF-CR&D SystemsCat # 9199-VC-025
Chemical compound, drugGefitinibCell Signaling TechnologyCat # 4765
Chemical compound, drugHydrocortisoneSigma-AldrichCat # H0888
Chemical compound, drugPenicillin/ streptomycinGibcoCat # 15140–122
Chemical compound, drugTrypLE ExpressGibcoCat # 12605-028
Chemical compound, drugFuGENE HDPromegaCat # E2311
Chemical compound, drugLipofectamine 3000Thermo Fisher ScientificCat # L3000015
Chemical compound, drugAminopropyl trimethoxysilaneSigma-AldrichCat # 281778
Chemical compound, drugGlutaraldehydeSigma-AldrichCat # 340855
Chemical compound, drug40% acrylamide solutionBio-RadCat # 1610140
Chemical compound, drug2% bis-acrylamide solutionBio-RadCat # 161-0142
Chemical compound, drugN,N,N’,N’-Tetramethyl ethylenediamine (TEMED)Sigma-AldrichCat # T9281
Chemical compound, drugAmmonium persulfate (APS)Sigma-AldrichCat # A3678
Commercial assay or kitinFusion HD cloning kitClonTech LaboratoriesCat # 638911Cloning kit
OtherDMEM/F12GibcoCat # 11320033Culture media
OtherHorse serumGibcoCat # 16050122Serum for culture media
OtherDMEMGibcoCat # 11995-065Culture media
OtherFetal bovine serumR&D SystemsCat # S11150Serum for culture media
Software, algorithmFIJISchindelin et al., 2012http://fiji.sc;
RRID:SCR_00228
Software, algorithmPython code for computational model; analysis code for raw dataThis paperhttps://github.com/toettchlab/Farahani2022/ (copy archived at toettchlab, 2023)
Software, algorithmR Studio 1.1.456RStudiorstudio.com; RRID:SCR_000432
Appendix 1—table 1
Equations used in the mathematical model.

L-EGFR: ligand-bound EGFR; EGFR:EGFR: EGFR in dimeric form; EGFR: EGFR bound to ZtSH2.

SpeciesNotationEquation
Soluble ligand (L)N1dN1dt=0 (A1)
Unbound ZtSH2 (*)N2dN2dt=k8N2(2N6+2N7+N9+N10+N11)+k9(N8+N9+N10+N11+2N12+2N13+2N14+N15+N16) (A2)
EGFRN3dN3dt=k1N3N1+k2N42k5N32k5N3N4k5N3N15k5N3N16+2k6N5+k6N8+k7N6+k7N9+k9N15 (A3)
L-EGFRN4dN4dt=k1N3N1k2N4k5N3N42k5N42k5N4N15k5N4N16+k7N6+k7N10+2k7N7+k7N11+k9N16 (A4)
EGFR:EGFRN5dN5dt=-2k1N5N1+k3N6+k5N32-k6N5+k9N8 (A5)
L-EGFR:EGFRN6dN6dt=k1N6N1+2k4N7+2k1N5N1k3N6+k5N3N4k7N62k8N6N2+k9N9+k9N10 (A6)
L-EGFR:L-EGFRN7dN7dt=k5N6N1-2k4N7+k5N42-k7N7-2k8N7N2+k9N11 (A7)
EGFR*:EGFRN8dN8dt=-2k1N8N1+k3N9+k3N10+k5N3N15-k6N8-k9N8+2k9N12 (A8)
L-EGFR*:EGFRN9dN9dt=k1N8N1-k1N9N1-k3N9+k4N11+k5N3N16-k7N9+k8N6N2-k8N9N2-k9N9+k9N13 (A9)
L-EGFR:EGFR*N10dN10dt=k1N8N1-k3N10-k1N10N1+k4N11+k5N4N15-k7N10+k8N6N2-k9N10-k8N10N2+k9N13 (A10)
L-EGFR*:L-EGFRN11dN11dt=k1N9N1-2k4N11+k1N10N1+k5N4N16-k7N11+2k8N7N2-k9N11-k8N11N2+2k9N14 (A11)
EGFR*:EGFR*N12dN12dt=-2k1N12N1+k3N13+k5N152-k6N12-2k9N12 (A12)
L-EGFR*:EGFR*N13dN13dt=2k1N12N1-k3N13-k1N13N1+2k4N14+k5N15N16-k7N13+k8N9N2-2k9N13+k8N10N2 (A13)
L-EGFR*:L-EGFR*N14dN14dt=k1N13N1-2k4N14+k5N162-k7N14+k8N11N2-2k9N14 (A14)
EGFR*N15dN15dt=-k1N15N1+k2N16-2k5N152+2k6N12-k5N3N15+k6N8-k5N15N16+k7N13-k5N4N15+k7N10-k9N15 (A15)
L-EGFR*N16dN16dt=k1N15N1-k2N16-2k5N162+2k7N14-k5N15N16+k7N13-k5N16N3+k7N9-k5N4N16+k7N11-k9N16 (A16)
Appendix 1—table 2
Parameters used in the mathematical model.
ParameterNotationValueUnitsNotes
Receptor–ligand bindingk10.03nM–1 s–1Schoeberl et al., 2002
Ligand dissociating from L-EGFR (β = 1 corresponds to EGF)k2β*6.6e-3s–1Macdonald and Pike, 2008
Ligand dissociating from L-EGFR:EGFR (β = 1 corresponds to EGF)k3β*5.7e-3s–1Macdonald and Pike, 2008
Ligand dissociating from L-EGFR:L-EGFRk40.087s–1Macdonald and Pike, 2008
Receptor dimerization and activationk51e-5nM–1 s–1Estimated
EGFR:EGFR dissociationk65e-3; variable values in Figure 3—figure supplement 2.s–1Estimated
L-EGFR:EGFR dissociation (γ = 1 corresponds to EGF)k7γ*1e-4s–1Estimated
ZtSH2 binding to receptork85nM–1 s–1Kd from Ottinger et al., 1998; kinetics set to be ~10 s based on our experimental measurements of ZtSH2 translocation
ZtSH2 dissociating from receptork916.67s–1Kd from Ottinger et al., 1998; kinetics set to be ~10 s based on our experimental measurements of ZtSH2 translocation
Scaling parameter for ligand–receptor bindingβ1 for EGF; 50 for low-affinity ligands; variable values in Figure 3CUnitlessFreed et al., 2017
Scaling parameter for dimerization of ligand-bound receptorsγ1 for EGF; 100 for low-affinity ligands; variable values in Figure 3CUnitlessFreed et al., 2017; 
Hu et al., 2022

Additional files

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Payam E Farahani
  2. Xiaoyu Yang
  3. Emily V Mesev
  4. Kaylan A Fomby
  5. Ellen H Brumbaugh-Reed
  6. Caleb J Bashor
  7. Celeste M Nelson
  8. Jared E Toettcher
(2023)
pYtags enable spatiotemporal measurements of receptor tyrosine kinase signaling in living cells
eLife 12:e82863.
https://doi.org/10.7554/eLife.82863