Cell-surface tethered promiscuous biotinylators enable comparative small-scale surface proteomic analysis of human extracellular vesicles and cells

  1. Lisa L Kirkemo
  2. Susanna K Elledge
  3. Jiuling Yang
  4. James R Byrnes
  5. Jeff E Glasgow
  6. Robert Blelloch
  7. James A Wells  Is a corresponding author
  1. Department of Pharmaceutical Chemistry, University of California, San Francisco, United States
  2. Department of Urology, University of California, San Francisco, United States
  3. The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, United States
  4. Department of Cellular and Molecular Pharmacology, University of California, San Francisco, United States
5 figures, 2 tables and 2 additional files

Figures

Figure 1 with 3 supplements
Direct labeling of promiscuous biotinylators to the cell membrane for rapid cell surface proteome characterization of small-scale biological samples.

(A) Outline of enzymatic reaction mechanism. APEX2 and HRP both require biotin tyramide and hydrogen peroxide to produce the biotin-radical intermediate. (B) Tethering either enzyme is completed through differing mechanisms: (i) APEX2 is tethered through bio-conjugation of a single-strand of DNA, which is complementary to an exogenously added sequence of lipidated-DNA attached to the membrane, (ii) Commercially available wheat germ agglutinin (WGA)-HRP associates with native GlcNAc and sialic acid glycan moieties on cell surface proteins. (C) Microscopy images depicting the localization of DNA-APEX2 to the cell surface of KP-4 cells after the introduction of the lipidated-DNA complementary strands. (D) Microscopy images depicting the localization of WGA-HRP to the membrane of KP-4 cells. All microscopy images are representative of two biological replicates.

Figure 1—figure supplement 1
Expression, purification, and validation of APEX2 enzyme.

His-tagged APEX2 was expressed in BL21(DE3)pLysS cells and purified by a nickel column. Ten milligrams of the purified enzyme were run out on a 4%–12% Bis-Tris gel to confirm purity.

Figure 1—figure supplement 2
Labeling and efficacy of APEX2 with DNA.

(A) APEX2 was first conjugated with DBCO-Maleimide (DBCO-Mal) reagent at 40 equivalents for 5 hr (80% conversion to the singly labeled product). Following desalting, three equivalences of Azide-DNA were added to the conjugate and purified by a Nickel column. Both reactions were monitored by LC-MS as shown. (B) 500,000 Expi293 cells were labeled with 0.5 µM purified APEX2 and DBCO-labeled APEX2 for 2 min. Extent of biotinylation of target cells was quantified by flow cytometry staining with streptavidin-647. (C) The DNA-APEX2 conjugate was shown to be tethered in the presence of the lipidated DNA (purple) and not in the absence (green), as detected by an Anti-His 680 antibody. Unlabeled APEX2 (blue) additionally did not result in a signal shift. Flow cytometry images are representative of one biological replicate.

Figure 1—figure supplement 3
WGA-HRP preincubation time on cells has no effect on labeling efficiency.

WGA-HRP was incubated on Expi293 cells for 0–30 min to determine optimal incubation time on ice before labeling. All tested times resulted in similar cell surface biotinylation efficiencies and signified that no incubation time was needed. Flow cytometry images are representative of one biological replicate.

Figure 2 with 6 supplements
Membrane-localized peroxidases increases membrane proteome biotinylation compared to nontethered counterparts.

(A) Biotinylation of Expi293 cells treated with free enzyme (APEX2 or HRP) or cell-tethered enzyme (DNA-APEX2 or WGA-HRP) shown by flow cytometry. Signal is read out using Streptavidin-AlexaFluor-647. (B) Comparison of cell labeling with either free enzyme (APEX2 or HRP) or cell-tethered enzyme (DNA-APEX2 or WGA-HRP) shown by Streptavidin-800 western blot and total protein stain. Normalized area is plotted to the right. (C) Number of cell membrane proteins identified by mass spectrometry (>2 unique peptides, <1% FDR, found in both biological replicates) after treating 500,000 KP-4 pancreatic cancer cells with either free enzyme (APEX2 or HRP) or cell-tethered enzyme (DNA-APEX2 or WGA-HRP). (D) Microscopy images depicting extent of labeling with free HRP compared to WGA-HRP with and without the blocking sugar GlcNAc. All western blot images, microscopy images, mass spectrometry data, and flow cytometry data are representative of two biological replicates.

Figure 2—figure supplement 1
Optimization of APEX2 concentrations on cell by flow cytometry.

(A) 500,000 Expi293 cells were labeled for 2 min with increasing amounts of purified APEX2 enzyme and extent of labeling was quantified by flow cytometry staining with streptavidin-647. (B) Varying numbers of Expi293 cells were labeled for 2 min with 0.5 µM APEX2 to test range of cell numbers for labeling. Flow cytometry images are representative of one biological replicate.

Figure 2—figure supplement 2
Rank ordered intensities for surface annotated proteins detected in tethered and untethered enzyme samples.

(A) A rank-ordered list of MS1 intensities (area) for surface annotated proteins found in APEX2 and DNA-APEX2 samples. (B) A rank-ordered list of MS1 intensities (area) for surface annotated proteins found in HRP and WGA-HRP samples. The average of two biological replicates is plotted.

Figure 2—figure supplement 3
Comparison of enrichment for glycosylated and non-glycosylated proteins.

Total MS1 intensities (LFQ area) of Uniprot annotated glycoproteins compared to non-glycoproteins in the SURFY database for each labeling method compared to the total area sum intensity. LFQ, label-free quantification.

Figure 2—figure supplement 4
Total plasma membrane (PM) protein identifications for DNA-APEX2 and WGA-HRP labeling experiments as a function of time.

500,000 PaTu8902 pancreatic cancer cells were labeled with either 0.5 µM DNA-APEX2 or 0.5 µM WGA-HRP for 1 or 2 min at 37°C. After cell surface enrichment and mass spectrometry analysis, the PM-derived protein identifications were totaled. Two biological replicates are plotted.

Figure 2—figure supplement 4—source data 1

Mass spectrometry analysis results table.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig2-figsupp4-data1-v2.csv
Figure 2—figure supplement 5
WGA-HRP labeling is N-acetyl-D-glucosamine (GlcNAc) dependent.

Biotinylation of RWPE-1 Myc cells with WGA-HRP was determined with (orange) and without (dark blue) 100 mg/ml GlcNAc. There is a significant leftward shift in the degree of labeling in the absence of competing GlcNAc, demonstrating that the enhanced labeling by WGA-HRP is GlcNAc dependent. The degree of labeling is similar to soluble HRP, as shown in light blue. Importantly, the presence of GlcNAc in solution did not generally affect HRP labeling as seen by the control in red. The data presented are from one biological replicate.

Figure 2—figure supplement 6
WGA-HRP can be used to label adherent cells on-plate.

Cell surface labeling was compared between labeling adherent cells on a tissue culture plate versus lifting cells and then performing labeling. Cell surface biotinylation was detected by streptavidin-Alexa Fluor 647. The data presented are from one biological replicate.

Figure 3 with 2 supplements
WGA-HRP identifies a number of enriched markers on Myc-driven prostate cancer cells.

(A) Overall scheme for biotin labeling, and label-free quantification (LFQ) by LC-MS/MS for RWPE-1 Control and Myc overexpression cells. (B) Microscopy image depicting morphological differences between RWPE-1 Control and RWPE-1 Myc cells after 3 days in culture. (C) Volcano plot depicting the LFQ comparison of RWPE-1 Control and Myc labeled cells. Red labels indicate upregulated proteins in the RWPE-1 Control cells over Myc cells and green labels indicate upregulated proteins in the RWPE-1 Myc cells over Control cells. All colored proteins are at least two-fold enriched in either dataset between four replicates (two technical, two biological, p<0.05). (D) Heatmap of the 30 most upregulated transmembrane (bold) or secreted (italics) proteins in either RWPE-1 Control or Myc cells. Scale indicates intensity, defined as (LFQ Area−Mean LFQ Area)/Standard Deviation. (E) Table indicating fold-change of most differentially regulated proteins by LC-MS/MS for RWPE-1 Control and Myc cells. (F) Upregulated proteins in RWPE-1 Myc cells (Myc, ANPEP, Vimentin, and FN1) are confirmed by western blot. (G) Upregulated surface proteins in RWPE-1 Myc cells (Vimentin, ANPEP, and FN1) are detected by immunofluorescence microscopy. The downregulated protein HLA-B by Myc overexpression was also detected by immunofluorescence microscopy. All western blot images and microscopy images are representative of two biological replicates. Mass spectrometry data is based on two biological and two technical replicates (N=4).

Figure 3—source data 1

Uncropped western blots.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig3-data1-v2.png
Figure 3—source data 2

Mass spectrometry analysis results table.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig3-data2-v2.xlsx
Figure 3—source data 3

List of proteins comparing enriched targets (>2-fold) in Myc cells versus Control cells.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig3-data3-v2.xlsx
Figure 3—figure supplement 1
Comparison of surface enrichment between replicates for different mass spectrometry methods.

(A) The top three methods (NHS-Biotin, Biocytin Hydrazide, and WGA-HRP) were compared for their ability to enrich cell surface proteins on 1.5 M RWPE-1 Control cells by LC-MS/MS after being searched with the Uniprot GOCC Plasma Membrane database. Shown are enrichment levels on the protein, peptide, and average MS1 intensity of top three peptides (LFQ area) levels. (B) The top three methods (NHS-Biotin, Biocytin Hydrazide, and WGA-HRP) were compared for their ability to enrich cell surface proteins on 1.5 M RWPE-1 Control cells by LC-MS/MS after being searched with the entire human Uniprot database. Shown are enrichment levels on the protein, peptide, and average MS1 intensity of top three peptides (LFQ area) levels. Proteins or peptides detected from cell surface annotated proteins (determined by the SURFY database) were divided by the total number of proteins or peptides detected. LFQ areas corresponding to cell surface annotated proteins (SURFY) were divided by the total area sum intensity for each sample. The corresponding percentages for two biological replicates were plotted. LFQ, label-free quantification.

Figure 3—figure supplement 2
Comparison of replicates for different mass spectrometry methods shows that WGA-HRP has comparable reproducibility to NHS-Biotin and Hydrazide labeling.

(A) Spearman correlations of total area sum intensity normalized data from replicates of Hydrazide Control and Myc cells. (B) Spearman correlations of total area sum intensity normalized data from replicates of NHS Control and Myc cells. (C) Spearman correlations of total area sum intensity normalized data from replicates of WGA Control and Myc cells.

Figure 4 with 1 supplement
WGA-HRP identifies a number of enriched markers on Myc-driven prostate cancer EVs.

(A) Workflow for small EV isolation from cultured cells. (B) Labeled proteins indicating canonical exosome markers (ExoCarta Top 100 List) detected after performing label-free quantification (LFQ) from whole EV lysate. The LFQ intensities were averaged for Control and Myc EVs, and the resulting protein list is graphed from least abundant to most abundant. (C) Workflow of EV labeling and preparation for mass spectrometry. (D) Heatmap of the 30 most upregulated proteins in either RWPE-1 Control or Myc EVs. Scale indicates intensity, defined as (LFQ Area−Mean LFQ Area)/Standard Deviation. (E) Table indicating fold-change of most differentially regulated proteins by LC-MS/MS for RWPE-1 Control and Myc cells. (F) Upregulated proteins in RWPE-1 Myc EVs (ANPEP and FN1) are confirmed by western blot. Mass spectrometry data is based on two biological and two technical replicates (N=4). Due to limited sample yield, one replicate was performed for the EV western blot. EV, extracellular vesicle.

Figure 4—source data 1

Uncropped western blots.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig4-data1-v2.png
Figure 4—source data 2

Whole EV mass spectrometry analysis results table.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig4-data2-v2.xlsx
Figure 4—source data 3

Mass spectrometry analysis results table.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig4-data3-v2.xlsx
Figure 4—source data 4

List of proteins comparing enriched targets (>1.5-fold) in Myc EVs versus Control EVs.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig4-data4-v2.xlsx
Figure 4—figure supplement 1
Venn diagram comparing enriched targets (>2-fold) in cells and EVs.

(A) Surface and secreted targets were found enriched in the Control EVs (purple) and Control cells (blue) when each is separately compared to Myc EVs and Myc cells, respectively. The four overlapping enriched targets in common between Control cells and Control EVs are listed in the center. (B) Surface and secreted targets were found enriched in the Myc EVs (purple) and Myc cells (blue) when each is separately compared to Control EVs and Control cells, respectively. The 12 overlapping enriched targets in common between Myc cells and Myc EVs are listed in the center.

Figure 5 with 1 supplement
WGA-HRP identifies a number of EV-specific markers that are present regardless of oncogene status.

(A) Matrix depicting samples analyzed during LFQ comparison–Control and Myc cells, as well as Control and Myc EVs. (B) Principle component analysis (PCA) of all four groups analyzed by LFQ. Component 1 (50.4%) and component 2 (15.8%) are graphed. (C) Functional annotation was performed for each gene cluster using DAVID Bioinformatics Resource 6.8 and the highest ranking annotation features for the EV-specific gene cluster are shown. (D) Heatmap of the 50 most upregulated proteins in either RWPE-1 cells or EVs. Proteins are listed in decreasing order of expression with the most highly expressed proteins in EVs on the far left and the most highly expressed proteins in cells on the far right. Averages from all four replicates of each sample type are graphed. Scale indicates intensity, defined as (LFQ Area−Mean LFQ Area)/Standard Deviation. Extracellular proteins with annotated transmembrane domains are bolded and annotated secreted proteins are italicized. (E) Table indicating fold-change of most differentially regulated proteins by LC-MS/MS for RWPE-1 EVs compared to parent cells. (F) Western blot showing the EV-specific marker ITIH4, IGSF8, and MFGE8. Mass spectrometry data is based on two biological and two technical replicates (N=4). Due to limited sample yield, one replicate was performed for the EV western blot. EV, extracellular vesicle; LFQ, label-free quantification.

Figure 5—source data 1

Uncropped western blots.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig5-data1-v2.png
Figure 5—source data 2

Mass spectrometry analysis results table.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig5-data2-v2.xlsx
Figure 5—source data 3

List of proteins comparing enriched targets (>2-fold) in Control EVs versus Control cells and Myc EVs versus Myc cells.

https://cdn.elifesciences.org/articles/73982/elife-73982-fig5-data3-v2.xlsx
Figure 5—figure supplement 1
Heatmap comparison of biological and technical replicates of RWPE-1 Control/Myc cells and EVs.

Biological and technical replicates cluster together based on both oncogene status and compartment for EV or cell surface. Proteins with no area values were assigned an imputed value using Perseus. Heatmap clustering is based off of the Pearson correlation between all replicates on both columns and rows. Heatmap was produced using Morpheus, https://software.broadinstitute.org/morpheus. The first number following the sample name denotes the biological replicated and second number denotes the technical replicate.

Tables

Table 1
Current methods available for cell surface biotinylation.
MethodProtocol length (time)SelectivitySample size requirement
Biocytin hydrazide+++++++++
Sulfo-NHS-LC-LC-biotin++++
APEX2/HRP+++
Key resources table
Reagent type (species)
or resource
DesignationSource or referenceIdentifiersAdditional information
Strain, strain background (Escherichia coli)BL21(DE3)pLysSPromegaL1195
Cell line (human)Expi293FThermo Fisher ScientificA14527
Cell line (human)PaTu8902https://doi.org/10.7554/eLife.45313.001
Cell line (human)KP-4https://doi.org/10.7554/eLife.45313.001
Cell line (human)RWPE-1https://doi.org/10.1073/pnas.2018861118Control and Myc overexpression lines
AntibodyAnti-HisTag-650 (mouse monoclonal)InvitrogenMA1-21315-D6501:100, flow cytometry, immunocytochemistry
AntibodyBiotin-conjugated anti-HRP (rabbit polyclonal)Rockland200-4638-01001:100, flow cytometry, immunocytochemistry
AntibodyAnti-ANPEP (sheep polyclonal)R&D SystemsAF38151:1000
AntibodyAnti-Vimentin (rabbit monoclonal)Cell Signaling Technology5741S1:1000
AntibodyAnti-FN1 (rabbit polyclonal)Abcamab24131:1,000
AntibodyAnti-HLA-B (rabbit polyclonal)ProteinTech17260-1-AP1:1,000
AntibodyAnti-ITIH4 (rabbit polyclonal)Atlas antibodiesHPA0039481:1,000
AntibodyAnti-MFGE8 (rabbit polyclonal)Thermo Fisher ScientificPA5-820361:1,000
AntibodyAnti-IGSF8 (goat polyclonal)R&D SystemsAF3117-SP1:1,000
AntibodyGoat Anti-Rabbit HRP (goat polyclonal)Thermo Fisher Scientific314601:10,000
AntibodyRabbit Anti-Sheep HRP (rabbit polyclonal)Thermo Fisher Scientific314801:10,000
Recombinant DNA reagentAPEX2Twist Biosciences
Sequence-based reagentDBCO-DNA (Conjugated to APEX2)IDT
Sequence-based reagentLipid DNA (Anchor and Co-Anchor)https://doi.org/10.1038/s41592-019-0433-8
Peptide, recombinant proteinHRP, PeroxidaseThermo Fisher Scientific31,490
Peptide, recombinant proteinWGA, Peroxidase (WGA-HRP)Vector LaboratoriesPL-1026-2
Peptide, recombinant proteinIRDye 800CW StreptavidinLi-Cor926-322301:10,000
Peptide, recombinant proteinIRDye 680RD Donkey anti-Goat IgGLi-Cor925-680741:10,000
Commercial assay or kitBCA assayThermo Fisher Scientific23228
Commercial assay or kitPreomics iST kitPreOmicsP.O.00027
Commercial assay or kitPierce quantitative colorimetric peptide assayThermo Fisher Scientific23275
Commercial assay or kitExpi293 mediaThermo Fisher ScientificA1435101
Commercial assay or kitKeratinocyte SFM media (1×)Thermo Fisher Scientific17005042
Commercial assay or kitMycoAlert PLUS mycoplasma detection kitLonzaLT07-703
Chemical compound, drugTroloxFisher Scientific501176131
Chemical compound, drugSodium AscorbateSigma-AldrichA4034-100G
Chemical compound, drugN-acetyl-D-glucosamineSigma-AldrichA3286-5G
Chemical compound, drugH2O2 (1×)Sigma-AldrichH1009-100ML
Chemical compound, drugHemin-ClSigma-Aldrich51280-1G
Chemical compound, drugMaleimide DBCOClick Chemistry ToolsA108P-10
Chemical compound, drugBiotin TyramideSigma-AldrichSML2135-50MG
Chemical compound, drugSodium PyruvateSigma-AldrichS8636-100ML
Chemical compound, drugSodium PeriodateSigma-Aldrich311448-5G
Chemical compound, drugBiocytin HydrazideBiotium90060
Chemical compound, drugAnilineSigma-Aldrich242284
Chemical compound, drugProtease Inhibitor CocktailSigma-AldrichP8340-1ML
Chemical compound, drugHigh Capacity Neutravidin ResinThermo Fisher Scientific29204
Chemical compound, drugPoly-D-lysineThermo Fisher ScientificA3890401
Chemical compound, drugSulfo NHS LC-LC BiotinThermo Fisher ScientificA35358
Software, algorithmFlowJoFlowJo (https://www.flowjo.com)RRID:SCR_008520
Software, algorithmGraphPad PrismPrism (https://www.graphpad.com/scientific-software/prism/)RRID:SCR_002798
Software, algorithmMorpheusMorpheus (https://software.broadinstitute.org/morpheus/)
Software, algorithmFIJIFIJI (https://fiji.sc)RRID:SCR_002285
OtherPenicillin/streptomycinThermo Fisher Scientific15-140-122
OtherFetal bovine serumGemini Bio-Products100-106
OtherTrypsin (0.05%)Life Technologies25300054

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. Lisa L Kirkemo
  2. Susanna K Elledge
  3. Jiuling Yang
  4. James R Byrnes
  5. Jeff E Glasgow
  6. Robert Blelloch
  7. James A Wells
(2022)
Cell-surface tethered promiscuous biotinylators enable comparative small-scale surface proteomic analysis of human extracellular vesicles and cells
eLife 11:e73982.
https://doi.org/10.7554/eLife.73982