2019 ASMS Adeptrix Poster FINAL 052919 · (—THIS SIDEBAR DOES NOT PRINT—) DESIGN GUIDE This...

1
Cell Culture, Serum & Tissue : MKN-45 and HeLa cells were prepared in RPMI media with 10% fetal bovine serum (FBS) and 1X Pen-Strep (Sigma, #P4333) to 75% confluence at 37 °C with 5% CO2. Prior to chemical treatment, cells were serum starved in RPMI media with 0.2% FBS and 1% Pen/Strep for 12 hrs. SU11274 (SU) and staurosporine (ST) were used at a final concentration of 1 μM and 0.2 μM, respectively in 0.05% DMSO. Hydrogen peroxide (H2O2) was used at a final concentration of 2 mM with a 30 m pre-treatment of 0.1 mM sodium orthovanadate. Rapamycin treatment was carried out for a duration of 2 hours at a concentration of 1 mM. Serum and samples were obtained from commercial sources. Human brain samples were obtained from the Maine Medical Research Institute Biobank repository. Preparation of Protein Lysates and Digested Peptides : Cells were washed twice with cold PBS. PBS was removed and cells were scraped in Urea Lysis Buffer (ULB, 8 M sequanal grade Urea, 20 mM HEPES pH 8.0, 1 mM β-glycerophosphate, 1 mM sodium vanadate, 2.5 mM sodium pyrophosphate). Tissue was pulverized under liquid nitrogen using a Bessman press, and transferred into ULB. The tissue slurry was homogenized using a mini-beadbeater. Cell cultures material and homogenized tissue were sonicated 3 times for 20 s each at 15 W output power with a 1-minute cooling on ice between each burst. Sonicated lysates were centrifuged 15 min at 4 °C at 20,000× g. An aliquot of each supernatant was reserved for Western blotting (if needed) and stored at 80 °C. Supernatants were collected and reduced with 4.5 mM DTT for 30 min at 40 °C. Reduced lysates were alkylated with 10mM iodoacetamide for 15 min at room temperature in the dark. Samples were diluted 1:4 with 0.2% ammonium bicarbonate (pH 8.0) and digested overnight with trypsin-TPCK (1:75, w:w, Promega) in 1 mM HCl. Other protease digestions were performed using the manufacturer’s recommended protocol (LysC, AspN, GluC, ArgC, Promega and NEB). Digested peptide lysates were desalted over 360 mg SEP PAK Classic C18 columns (Waters, Richmond, VA, USA, #WAT051910). Peptides were eluted with 50% acetonitrile in 0.1% TFA, dried under lyophilization conditions, and stored at 80 °C in 0.1 – 1.0 mg aliquots. Immunoaffinity Enrichment & MALDI Analysis : Protein A/G beads were prepared using NHS-activated XL magnetic agarose beads (400 micron, Cube Biotech) with Protein A/G (Abcam, 1 mg/ml) in PBS buffer. Antibodies (2 µg) were conjugated to 5 µL slurry of Protein A/G beads by overnight incubation in PBS with 0.1% BSA. Unbound antibody was removed with three 400 µL washes of PBS with 0.1% BSA. Individual target peptide enrichment was performed using 40 – 1000 µg of purified peptides with 1 – 3 beads. Multiplex target peptide enrichment was performed using 40 – 1000 µg of purified peptides with 3 beads per protein target. Peptides were incubated overnight at 4 °C. Beads were washed three times in PBS to remove nonspecific bound peptides. Stringent wash conditions included PBS with 0.5 – 1.0 M sodium chloride. Final washes included 10 mM ammonium bicarbonate (pH 7.5) and distilled water (MilliQ). Washed beads were transferred to ITO (or gold) slides affixed with the pico- well gasket and a sample gasket. Pico-wells were hydrated prior to adding washed beads using a benchtop swinging bucket centrifuge at low speed (~ 1000 xg). For single bead analysis, all liquid was removed from last wash and add 1.5 – 2.0 µL of matrix (10 mg/mL CHCA in 50% ethanol/water, 0.1% formic acid) to elute bound peptide(s) for 15 min at 25 °C. Spot 1uL of eluted peptides (in matrix) onto the MALDI plate. Allow to dry completely before MS analysis using a MALDI TOF instrument (Autoflex Speed, Bruker & SimulTOF ONE, SimulTOF). INTRODUCTION METHODS Proteomic studies that monitor protein and PTM abundance often employ multi-dimensional analytical methods such as nano-LC-ESI-MS/MS to simplify the inherent sample complexity and wide dynamic range of endogenous proteins within biological specimens. The time and expertise required to implement and LCMS workflow can often be a barrier to integrating targeted proteomic applications for a particular translational research program. In addition, sample quantity requirements limit accessibility of LCMS-based targeted methods as a practical screening platform. In this study, we present a versatile microarray assay platform (BAMS TM ) that integrates immuno-affinity capture with MALDI MS detection, which can be leveraged to perform both in vivo targeted proteomic assays as well as in vitro enzyme- substrate assays for a wide range of high-throughput screening applications. METHODS METHODS & RESULTS CONCLUSIONS Multiplexing of BAMS enables one to monitor 100’s of proteins in a single assay. The versatility of the BAMS allows rapid configuration of targeted assays monitoring many aspects of the protein (unmodified, phospho-, acetyl- &, methyl-). BAMS assays are an efficient method to monitor a wide variety of proteins in any type of biological samples. BAMS assays can be utilized to perform a wide range of targeted proteomic applications and is amenable for high-throughput screening. 1US patent 9,618,520 by inventor V.Bergo, titled Devices and methods for producing and analyzing microarrays 2US patent 10,101,336 by inventor V.Bergo, titled Eluting analytes from bead arrays 3US patent application 16/125164 by inventor V.Bergo, titled Multiplexed bead arrays for proteomics ACKNOWLEDGEMENTS North Shore InnoVentures (https://nsiv.org/) and its corporate sponsor sand the Massachusetts Life Science Center (http://www.masslifesciences.com/) for grant support. Figure 1. Sample Preparation Workflow. Standard bottom-up methods are used to generate proteolytic peptides for subsequent BAMS analysis (lysis, reduction, alkylation, digestion). Purified peptides are incubated overnight with BAMS affinity capture beads in eppindorf tube or 96-well microtiter plate (1). Magnetic agarose beads are transferred, sequentially into wash buffers (PBS, ammonium bicarbonate, DDW) before placed into the BAMS slide (2). Washed beads are transferred into hydrated wells of BAMS chip with gentle agitation and a short centrifugation to settle beads into pico-wells (3), captured peptides from each bead are eluted into the pico-well using a matrix sprayer (4), eluted, dry peptides are analyzed after disassembling gaskets and placing into slide adapter for MALDI MS measurement (5). RESULTS Sergey Mamaev , Jeffrey C. Silva , Camilla Worsfold & Vladislav B. Bergo Adeptrix Corpora/on, Beverly, MA 01915 A HighThroughput MulJplexed Assay PlaNorm for Monitoring Protein Abundance in 96Well Cell Cultures or Product Profiles from EnzymeSubstrate ReacJons Figure 2. Apparatus & Components for BAMS Assay. The BAMS assay components include: ITO or gold slides, pico-well gasket, sample chamber gaskets, clamps and centrifuge adapter (A). Antibody beads are provided separately. The matrix sprayer provides optimized elution conditions for MALDI MS measurement (B). Eluted peptides on ITO BAMS slides for low, medium and high-density assays (C). Slide adapter for BAMS slide and standard MALDI slide (D). Fluorescent labeled peptide on bead (E) & eluted peptide in pico-well (F). Jeffrey C. Silva Adeptrix Corporation 100 Cummings Center, Suite 438Q Beverly, MA 01915 [email protected] www.adeptrix.com RESULTS Figure 5. Forward and Reverse Curves for the PCI Peptide in Normal Human Serum using BAMS. MALDI MS peptide signal from BAMS assay of PCI & sTfR in normal human serum (reflector mode). The endogenous concentration of the PCI peptide is determined by the plateau shown by the forward dilution curve (FWD) and the analytical sensitivity of the assay is revealed by the reverse dilution curve (REV) when conducted using SIS standards in triplicate. The average %CV for the analytical replicates was deteremimed to be 15% and the dynamic range for the PCI peptide spanned approximately 3 orders of magnitude. Figure 4. BAMS assay from Petri Dish and 96-Well Plate Cultures. MALDI MS signal from single bead peptide elution onto a BAMS assay slide for 4EBP1 (total), 4EBP1 (T37 & T46), AKT (total), A - F. Petri dish samples (A - C) were harvested from a SILAC experiment (light = control, heavy = peroxide, 250 µg total peptides), and BAMS assays were performed with medium density pico-well gasket (500 µm diameter wells, 400 µm beads). BAMS assays were conducted using peptides from 96-well cell culture (D - F) were performed with high density pico-well gasket (250 µm diameter wells, 200 µm beads, 10 µg total peptides). Figure 6. BAMS assay to Monitor PTM Status. (Unmodified & Phosphorylated proteins as well as WT & MT isoforms). MALDI MS signal from a BAMS assay for unmodified and phosphorylated (S101) 4EBP1 (A) as well as singly and doubly phosphorylated 4EBP1 (T37 & T46) with 1 (BLUE) and 2 (RED) missed cleavages (B) from peroxide treated MKN45 cells. Wild-type and point mutant (A107V) for 4EBP1 from HCT116 cells (C & D). Figure 7. BAMS assay to Monitor Protein Acetylation & Isoforms. Reflector mode MALDI MS signal from BAMS assays for VDAC1 & VDAC2. The captured N-terminal peptides to VDAC1 (A) and VDAC2 (B) were generated from LysC digested cells (8 cell line mix). The VDAC1 peptide, (M) A(Acetyl)VPPTYADLGK (S) (amino acids 2-12, calc. MH+ = 1173.615), and the VDAC2 peptide, (M) A(Acetyl)THGQTC ARPMC IPPSYADLGK (S) (amino acids 2-23, calc. MH+ = 2473.142), are both N-terminally processed with loss of methionine and addition of N-terminal acetylation. Figure 9. Monitor Multiple Sites of a Single Protein using BAMS. Multiple sites within a single protein can be monitored in a multiplex BAMS assay using validated antibodies to different regions of the target protein. An example is shown using three different affinity capture beads to 4EBP1, with the typical tryptic peptides highlighted in yellow and orange. Targeted peptide regions can be adjusted by using a different protease as illustrated above for LysC. Figure 8. BAMS assay to Monitor Multiplexed Ubiquitination Reactions. Black trace - MALDI signal from a BSA digest spiked with 6 K-ε-GG containing peptides at 1.25µM each. None of the peptides were identified. Red trace – MALDI signal from BSA / K-ε-GG containing peptides after incubation with anti- K-ε-GG BAMS beads for 2 hours, washing with ammonium bicarbonate and eluting captured peptides from K-ε-GG affinity beads onto MALDI slide. Five out of six K-ε-GG peptides were identified after BAMS assay (green box). Figure 3. BAMS Assay Validation Workflow. The specificity of affinity beads are validated by single bead affinity capture and localized peptide elution into the pico-well for MALDI MS and/or MS/MS to identify captured peptide(s). A MALDI MS spectral library is generated for each protease digestion condition and each Affi-BAMS bead reagent (A). The BAMS assay can accommodate thousands of target peptides (unmodified & protein PTM) in a single experiment on a slide for identification and quantification of the target proteins in the configured assay panel (B). a) trypsin b) chymotrypsin c) othersB A A B D E F C ~15 fmoles ~ 3 Orders of Magnitude Dynamic Range PCI peptide: EDQYHYLLDR sTfR, GFVEPDHYVVGAQR PCI (PAI3), EDQYHYLLDR M. Razavi et al, Clin Chem (2013) 59(10): 1514-1522. UNMODIFIED & PHOSPHORYLATED WILD-TYPE POINT MUTANT (A107V) SINGLE & DOUBLE PHOSPHORYLATION RAGGESSQFEMDI RAGGESSQFEMDI RVGGESSQFEMDI RNSPEDKRAGGESSQFEMDI Δ80 Δ28 Δ80 Δ80 A B RVVLGDGVQLPPGDYSTTPGGTLFSTTPGGTR C D RVVLGDGVQLPPGDYSTTPGGTLFSTTPGGTR FLMECRNSPVTKTPPR RAGGEESQFEMDI 1. Enrich 2. Wash 3. Assemble Bead Array 4. Elute Target PepJdes 5. Scan BAMS Chip microarray (bead array) MALDI scanner As li[le as 10 μg of total soluble protein Overlay of 28-Plex BAMS Assay Individual MALDI MS from Replicate BAMS assays Petri Dish (10cm) 96-Well Plate RAGGESSQFEMDI MH+ = 1468.637, Δ10 NSPEDKRAGGESSQFEMDI MH+ = 2138.929, Δ18 Δ10 Δ20 Δ10 RPHFPQFSYSASGTA MH+ = 1652.782, Δ10 VVLGDGVQLPPGDYSTTPGGTLFSTTPGGTR, MH+ = 3207.465 RVVLGDGVQLPPGDYSTTPGGTLFSTTPGGTR, MH+ = 3363.566 A B C D E F Ave FC = -1.1 Ave FC = 1.6 Ave FC = 1.0 J. E. Rodriguez et al, Curr Hypertens Rep (2009) 11(6): 396-405. K-ε-GG Enrichment & MALDI MS Trypsin Digestion E2 E1 E3 in vitro substrate proteins Ubiquitin Ligase Ubiquitinated substrates A B VDAC1: A(Acetyl)VPPTYADLGK VDAC2: A(Acetyl)THGQTC ARPMC IPPSYADLGK

Transcript of 2019 ASMS Adeptrix Poster FINAL 052919 · (—THIS SIDEBAR DOES NOT PRINT—) DESIGN GUIDE This...

Page 1: 2019 ASMS Adeptrix Poster FINAL 052919 · (—THIS SIDEBAR DOES NOT PRINT—) DESIGN GUIDE This PowerPoint 2007 template produces a 42”x90” presentation poster. You can use it

(—THIS SIDEBAR DOES NOT PRINT—) D E S I G N G U I D E

This PowerPoint 2007 template produces a 42”x90” presentation poster. You can use it to create your research poster and save valuable time placing titles, subtitles, text, and graphics.

We provide a series of online answer your poster production questions. To view our template tutorials, go online to PosterPresentations.com and click on HELP DESK.

When you are ready to print your poster, go online to PosterPresentations.com

Need assistance? Call us at 1.510.649.3001

Q U I C K S T A R T

Zoom in and out As you work on your poster zoom in and out to the level that is more comfortable to you. Go to VIEW > ZOOM.

Title, Authors, and Affiliations

Start designing your poster by adding the title, the names of the authors, and the affiliated institutions. You can type or paste text into the provided boxes. The template will automatically adjust the size of your text to fit the title box. You can manually override this feature and change the size of your text.

T I P : The font size of your title should be bigger than your name(s) and institution name(s).

Adding Logos / Seals Most often, logos are added on each side of the title. You can insert a logo by dragging and dropping it from your desktop, copy and paste or by going to INSERT > PICTURES. Logos taken from web sites are likely to be low quality when printed. Zoom it at 100% to see what the logo will look like on the final poster and make any necessary adjustments.

T I P : See if your company’s logo is available on our free poster templates page.

Photographs / Graphics You can add images by dragging and dropping from your desktop, copy and paste, or by going to INSERT > PICTURES. Resize images proportionally by holding down the SHIFT key and dragging one of the corner handles. For a professional-looking poster, do not distort your images by enlarging them disproportionally.

Image Quality Check Zoom in and look at your images at 100% magnification. If they look good they will print well.

ORIGINAL   DISTORTED  

Corner  handles  

Good

 prin

/ng  qu

ality

 

Bad  prin/n

g  qu

ality

 

Q U I C K S TA R T ( c o n t . )

How to change the template color theme You can easily change the color theme of your poster by going to the DESIGN menu, click on COLORS, and choose the color theme of your choice. You can also create your own color theme. You can also manually change the color of your background by going to VIEW > SLIDE MASTER. After you finish working on the master be sure to go to VIEW > NORMAL to continue working on your poster.

How to add Text The template comes with a number of pre-formatted placeholders for headers and text blocks. You can add more blocks by copying and pasting the existing ones or by adding a text box from the HOME menu.

Text size

Adjust the size of your text based on how much content you have to present. The default template text offers a good starting point. Follow the conference requirements.

How to add Tables

To add a table from scratch go to the INSERT menu and click on TABLE. A drop-down box will help you select rows and columns.

You can also copy and a paste a table from Word or another PowerPoint document. A pasted table may need to be re-formatted by RIGHT-CLICK > FORMAT SHAPE, TEXT BOX, Margins.

Graphs / Charts You can simply copy and paste charts and graphs from Excel or Word. Some reformatting may be required depending on how the original document has been created.

How to change the column configuration RIGHT-CLICK on the poster background and select LAYOUT to see the column options available for this template. The poster columns can also be customized on the Master. VIEW > MASTER.

How to remove the info bars

If you are working in PowerPoint for Windows and have finished your poster, save as PDF and the bars will not be included. You can also delete them by going to VIEW > MASTER. On the Mac adjust the Page-Setup to match the Page-Setup in PowerPoint before you create a PDF. You can also delete them from the Slide Master.

Save your work Save your template as a PowerPoint document. For printing, save as PowerPoint or “Print-quality” PDF.

Student discounts are available on our Facebook page. Go to PosterPresentations.com and click on the FB icon.

©  2015  PosterPresenta/ons.com  2117  Fourth  Street  ,  Unit  C                  Berkeley  CA  94710  [email protected]  

Cell Culture, Serum & Tissue: MKN-45 and HeLa cells were prepared in RPMI media with 10% fetal bovine serum (FBS) and 1X Pen-Strep (Sigma, #P4333) to 75% confluence at 37 °C with 5% CO2. Prior to chemical treatment, cells were serum starved in RPMI media with 0.2% FBS and 1% Pen/Strep for 12 hrs. SU11274 (SU) and staurosporine (ST) were used at a final concentration of 1 μM and 0.2 μM, respectively in 0.05% DMSO. Hydrogen peroxide (H2O2) was used at a final concentration of 2 mM with a 30 m pre-treatment of 0.1 mM sodium orthovanadate. Rapamycin treatment was carried out for a duration of 2 hours at a concentration of 1 mM. Serum and samples were obtained from commercial sources. Human brain samples were obtained from the Maine Medical Research Institute Biobank repository.

Preparation of Protein Lysates and Digested Peptides: Cells were washed twice with cold PBS. PBS was removed and cells were scraped in Urea Lysis Buffer (ULB, 8 M sequanal grade Urea, 20 mM HEPES pH 8.0, 1 mM β-glycerophosphate, 1 mM sodium vanadate, 2.5 mM sodium pyrophosphate). Tissue was pulverized under liquid nitrogen using a Bessman press, and transferred into ULB. The tissue slurry was homogenized using a mini-beadbeater. Cell cultures material and homogenized tissue were sonicated 3 times for 20 s each at 15 W output power with a 1-minute cooling on ice between each burst. Sonicated lysates were centrifuged 15 min at 4 °C at 20,000× g. An aliquot of each supernatant was reserved for Western blotting (if needed) and stored at −80 °C. Supernatants were collected and reduced with 4.5 mM DTT for 30 min at 40 °C. Reduced lysates were alkylated with 10mM iodoacetamide for 15 min at room temperature in the dark. Samples were diluted 1:4 with 0.2% ammonium bicarbonate (pH 8.0) and digested overnight with trypsin-TPCK (1:75, w:w, Promega) in 1 mM HCl. Other protease digestions were performed using the manufacturer’s recommended protocol (LysC, AspN, GluC, ArgC, Promega and NEB). Digested peptide lysates were desalted over 360 mg SEP PAK Classic C18 columns (Waters, Richmond, VA, USA, #WAT051910). Peptides were eluted with 50% acetonitrile in 0.1% TFA, dried under lyophilization conditions, and stored at −80 °C in 0.1 – 1.0 mg aliquots.

Immunoaffinity Enrichment & MALDI Analysis: Protein A/G beads were prepared using NHS-activated XL magnetic agarose beads (400 micron, Cube Biotech) with Protein A/G (Abcam, 1 mg/ml) in PBS buffer. Antibodies (2 µg) were conjugated to 5 µL slurry of Protein A/G beads by overnight incubation in PBS with 0.1% BSA. Unbound antibody was removed with three 400 µL washes of PBS with 0.1% BSA. Individual target peptide enrichment was performed using 40 – 1000 µg of purified peptides with 1 – 3 beads. Multiplex target peptide enrichment was performed using 40 – 1000 µg of purified peptides with 3 beads per protein target. Peptides were incubated overnight at 4 °C. Beads were washed three times in PBS to remove nonspecific bound peptides. Stringent wash conditions included PBS with 0.5 – 1.0 M sodium chloride. Final washes included 10 mM ammonium bicarbonate (pH 7.5) and distilled water (MilliQ). Washed beads were transferred to ITO (or gold) slides affixed with the pico-well gasket and a sample gasket. Pico-wells were hydrated prior to adding washed beads using a benchtop swinging bucket centrifuge at low speed (~ 1000 xg). For single bead analysis, all liquid was removed from last wash and add 1.5 – 2.0 µL of matrix (10 mg/mL CHCA in 50% ethanol/water, 0.1% formic acid) to elute bound peptide(s) for 15 min at 25 °C. Spot 1uL of eluted peptides (in matrix) onto the MALDI plate. Allow to dry completely before MS analysis using a MALDI TOF instrument (Autoflex Speed, Bruker & SimulTOF ONE, SimulTOF).

INTRODUCTION   METHODS  Proteomic studies that monitor protein and PTM abundance often employ multi-dimensional analytical methods such as nano-LC-ESI-MS/MS to simplify the inherent sample complexity and wide dynamic range of endogenous proteins within biological specimens. The time and expertise required to implement and LCMS workflow can often be a barrier to integrating targeted proteomic applications for a particular translational research program. In addition, sample quantity requirements limit accessibility of LCMS-based targeted methods as a practical screening platform. In this study, we present a versatile microarray assay platform (BAMSTM) that integrates immuno-affinity capture with MALDI MS detection, which can be leveraged to perform both in vivo targeted proteomic assays as well as in vitro enzyme-substrate assays for a wide range of high-throughput screening applications.

METHODS  

METHODS  &  RESULTS  

CONCLUSIONS  •  Multiplexing of BAMS enables one to monitor 100’s of proteins in a single assay. •  The versatility of the BAMS allows rapid configuration of targeted assays

monitoring many aspects of the protein (unmodified, phospho-, acetyl- &, methyl-). •  BAMS assays are an efficient method to monitor a wide variety of proteins in any

type of biological samples. •  BAMS assays can be utilized to perform a wide range of targeted proteomic

applications and is amenable for high-throughput screening.

1US patent 9,618,520 by inventor V.Bergo, titled Devices and methods for producing and analyzing microarrays 2US patent 10,101,336 by inventor V.Bergo, titled Eluting analytes from bead arrays 3US patent application 16/125164 by inventor V.Bergo, titled Multiplexed bead arrays for proteomics

ACKNOWLEDGEMENTS  •  North Shore InnoVentures (https://nsiv.org/) and its corporate sponsor sand the Massachusetts

Life Science Center (http://www.masslifesciences.com/) for grant support.

Figure 1. Sample Preparation Workflow. Standard bottom-up methods are used to generate proteolytic peptides for subsequent BAMS analysis (lysis, reduction, alkylation, digestion). Purified peptides are incubated overnight with BAMS affinity capture beads in eppindorf tube or 96-well microtiter plate (1). Magnetic agarose beads are transferred, sequentially into wash buffers (PBS, ammonium bicarbonate, DDW) before placed into the BAMS slide (2). Washed beads are transferred into hydrated wells of BAMS chip with gentle agitation and a short centrifugation to settle beads into pico-wells (3), captured peptides from each bead are eluted into the pico-well using a matrix sprayer (4), eluted, dry peptides are analyzed after disassembling gaskets and placing into slide adapter for MALDI MS measurement (5).

RESULTS  

Sergey  Mamaev  ,  Jeffrey  C.  Silva,  Camilla  Worsfold  &  Vladislav  B.  Bergo  Adeptrix  Corpora/on,  Beverly,  MA  01915  

A  High-­‐Throughput  MulJplexed  Assay  PlaNorm  for  Monitoring  Protein  Abundance  in  96-­‐Well  Cell  Cultures  or  Product  Profiles  from  Enzyme-­‐Substrate  ReacJons  

Figure 2. Apparatus & Components for BAMS Assay. The BAMS assay components include: ITO or gold slides, pico-well gasket, sample chamber gaskets, clamps and centrifuge adapter (A). Antibody beads are provided separately. The matrix sprayer provides optimized elution conditions for MALDI MS measurement (B). Eluted peptides on ITO BAMS slides for low, medium and high-density assays (C). Slide adapter for BAMS slide and standard MALDI slide (D). Fluorescent labeled peptide on bead (E) & eluted peptide in pico-well (F).

Jeffrey C. Silva Adeptrix Corporation

100 Cummings Center, Suite 438Q Beverly, MA 01915 [email protected] www.adeptrix.com

RESULTS  

Figure 5. Forward and Reverse Curves for the PCI Peptide in Normal Human Serum using BAMS. MALDI MS peptide signal from BAMS assay of PCI & sTfR in normal human serum (reflector mode). The endogenous concentration of the PCI peptide is determined by the plateau shown by the forward dilution curve (FWD) and the analytical sensitivity of the assay is revealed by the reverse dilution curve (REV) when conducted using SIS standards in triplicate. The average %CV for the analytical replicates was deteremimed to be 15% and the dynamic range for the PCI peptide spanned approximately 3 orders of magnitude.

Figure 4. BAMS assay from Petri Dish and 96-Well Plate Cultures. MALDI MS signal from single bead peptide elution onto a BAMS assay slide for 4EBP1 (total), 4EBP1 (T37 & T46), AKT (total), A - F. Petri dish samples (A - C) were harvested from a SILAC experiment (light = control, heavy = peroxide, 250 µg total peptides), and BAMS assays were performed with medium density pico-well gasket (500 µm diameter wells, 400 µm beads). BAMS assays were conducted using peptides from 96-well cell culture (D - F) were performed with high density pico-well gasket (250 µm diameter wells, 200 µm beads, 10 µg total peptides).

Figure 6. BAMS assay to Monitor PTM Status. (Unmodified & Phosphorylated proteins as well as WT & MT isoforms). MALDI MS signal from a BAMS assay for unmodified and phosphorylated (S101) 4EBP1 (A) as well as singly and doubly phosphorylated 4EBP1 (T37 & T46) with 1 (BLUE) and 2 (RED) missed cleavages (B) from peroxide treated MKN45 cells. Wild-type and point mutant (A107V) for 4EBP1 from HCT116 cells (C & D).

Figure 7. BAMS assay to Monitor Protein Acetylation & Isoforms. Reflector mode MALDI MS signal from BAMS assays for VDAC1 & VDAC2. The captured N-terminal peptides to VDAC1 (A) and VDAC2 (B) were generated from LysC digested cells (8 cell line mix). The VDAC1 peptide, (M) A(Acetyl)VPPTYADLGK (S) (amino acids 2-12, calc. MH+ = 1173.615), and the VDAC2 peptide, (M) A(Acetyl)THGQTCARPMCIPPSYADLGK (S) (amino acids 2-23, calc. MH+ = 2473.142), are both N-terminally processed with loss of methionine and addition of N-terminal acetylation.

Figure 9. Monitor Multiple Sites of a Single Protein using BAMS. Multiple sites within a single protein can be monitored in a multiplex BAMS assay using validated antibodies to different regions of the target protein. An example is shown using three different affinity capture beads to 4EBP1, with the typical tryptic peptides highlighted in yellow and orange. Targeted peptide regions can be adjusted by using a different protease as illustrated above for LysC.

Figure 8. BAMS assay to Monitor Multiplexed Ubiquitination Reactions. Black trace -MALDI signal from a BSA digest spiked with 6 K-ε-GG containing peptides at 1.25µM each. None of the peptides were identified. Red trace – MALDI signal from BSA / K-ε-GG containing peptides after incubation with anti- K-ε-GG BAMS beads for 2 hours, washing with ammonium bicarbonate and eluting captured peptides from K-ε-GG affinity beads onto MALDI slide. Five out of six K-ε-GG peptides were identified after BAMS assay (green box).

Figure 3. BAMS Assay Validation Workflow. The specificity of affinity beads are validated by single bead affinity capture and localized peptide elution into the pico-well for MALDI MS and/or MS/MS to identify captured peptide(s). A MALDI MS spectral library is generated for each protease digestion condition and each Affi-BAMS bead reagent (A). The BAMS assay can accommodate thousands of target peptides (unmodified & protein PTM) in a single experiment on a slide for identification and quantification of the target proteins in the configured assay panel (B).

magnetic beads

a) trypsin b) chymotrypsin c) others…

B  A  

A B D

E F

C

~15 fmoles

~ 3 Orders of Magnitude Dynamic Range

PCI peptide: EDQYHYLLDR

sTfR, GFVEPDHYVVGAQR

PCI (PAI3), EDQYHYLLDR

M. Razavi et al, Clin Chem (2013) 59(10): 1514-1522.

UNMODIFIED & PHOSPHORYLATED WILD-TYPE

POINT MUTANT (A107V) SINGLE & DOUBLE PHOSPHORYLATION

RAGGESSQFEMDI

RAGGESSQFEMDI RVGGESSQFEMDI

RNSPEDKRAGGESSQFEMDI

Δ80

Δ28

Δ80 Δ80

❶ ❷ ❶

A

B RVVLGDGVQLPPGDYSTTPGGTLFSTTPGGTR

C

D

RVVLGDGVQLPPGDYSTTPGGTLFSTTPGGTR FLMECRNSPVTKTPPR RAGGEESQFEMDI

1.  Enrich  

2.  Wash  

3.  Assemble  Bead  Array   4.  Elute  Target  PepJdes  

5.  Scan  BAMS  Chip  

microarray (bead array) MALDI scanner

As  li[le  as  10  µg  of  total  soluble  

protein  

Overlay of 28-Plex BAMS Assay

Individual MALDI MS from Replicate BAMS assays

Petri Dish (10cm) 96-Well Plate

RAGGESSQFEMDI MH+ = 1468.637, Δ10

NSPEDKRAGGESSQFEMDI MH+ = 2138.929, Δ18

Δ10 Δ20

Δ10

RPHFPQFSYSASGTA MH+ = 1652.782, Δ10

VVLGDGVQLPPGDYSTTPGGTLFSTTPGGTR, MH+ = 3207.465 RVVLGDGVQLPPGDYSTTPGGTLFSTTPGGTR, MH+ = 3363.566

A

B

C

D

E

F

Ave FC = -1.1

Ave FC = 1.6

Ave FC = 1.0

J. E. Rodriguez et al, Curr Hypertens Rep (2009) 11(6): 396-405.

K-ε-GG Enrichment

& MALDI MS

Trypsin Digestion E2 E1 E3

in vitro substrate proteins

Ubiquitin Ligase

Ubiquitinated substrates

A B VDAC1: A(Acetyl)VPPTYADLGK VDAC2: A(Acetyl)THGQTCARPMCIPPSYADLGK