DOE PAGES title logo U.S. Department of Energy
Office of Scientific and Technical Information

Title: Automated Coupling of Nanodroplet Sample Preparation with Liquid Chromatography–Mass Spectrometry for High-Throughput Single-Cell Proteomics

Abstract

Single-cell proteomics can provide critical biological insight into the cellular heterogeneity that is masked by bulk-scale analysis. Here, we have developed a nanoPOTS (nanodroplet processing in one pot for trace samples) platform and demonstrated its broad applicability for single-cell proteomics. However, because of nanoliter-scale sample volumes, the nanoPOTS platform is not compatible with automated LC-MS systems, which significantly limits sample throughput and robustness. To address this challenge, we have developed a nanoPOTS autosampler allowing fully automated sample injection from nanowells to LC-MS systems. We also developed a sample drying, extraction, and loading workflow to enable reproducible and reliable sample injection. The sequential analysis of 20 samples containing 10 ng tryptic peptides demonstrated high reproducibility with correlation coefficients of >0.995 between any two samples. The nanoPOTS autosampler can provide analysis throughput of 9.6, 16, and 24 single cells per day using 120, 60, and 30 min LC gradients, respectively. As a demonstration for single-cell proteomics, the autosampler was first applied to profiling protein expression in single MCF10A cells using a label-free approach. At a throughput of 24 single cells per day, an average of 256 proteins was identified from each cell and the number was increased to 731 when the Matchmore » Between Runs algorithm of MaxQuant was used. Using a multiplexed isobaric labeling approach (TMT-11plex), ~77 single cells could be analyzed per day. We analyzed 152 cells from three acute myeloid leukemia cell lines, resulting in a total of 2558 identified proteins with 1465 proteins quantifiable (70% valid values) across the 152 cells. These data showed quantitative single-cell proteomics can cluster cells to distinct groups and reveal functionally distinct differences.« less

Authors:
 [1];  [1]; ORCiD logo [2]; ORCiD logo [2]; ORCiD logo [2];  [2];  [2]; ORCiD logo [2]; ORCiD logo [3]; ORCiD logo [1];  [4]; ORCiD logo [1]
  1. Pacific Northwest National Lab. (PNNL), Richland, WA (United States). Environmental Molecular Sciences Lab. (EMSL)
  2. Pacific Northwest National Lab. (PNNL), Richland, WA (United States). Biological Sciences Division
  3. Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99354 United States
  4. Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354 United States; Brigham Young Univ., Provo, UT (United States). Dept. of Chemistry and Biochemistry
Publication Date:
Research Org.:
Pacific Northwest National Lab. (PNNL), Richland, WA (United States)
Sponsoring Org.:
USDOE Laboratory Directed Research and Development (LDRD) Program; USDOE Office of Science (SC), Biological and Environmental Research (BER); National Institutes of Health (NIH)
OSTI Identifier:
1673582
Report Number(s):
PNNL-SA-152553
Journal ID: ISSN 0003-2700
Grant/Contract Number:  
AC05-76RL01830; GM103493; U24CA210955
Resource Type:
Accepted Manuscript
Journal Name:
Analytical Chemistry
Additional Journal Information:
Journal Volume: 92; Journal Issue: 15; Journal ID: ISSN 0003-2700
Publisher:
American Chemical Society (ACS)
Country of Publication:
United States
Language:
English
Subject:
37 INORGANIC, ORGANIC, PHYSICAL, AND ANALYTICAL CHEMISTRY; nanoPOTS; proteomics; LC-MS; automation

Citation Formats

Williams, Sarah M., Liyu, Andrey V., Tsai, Chia-Feng, Moore, Ronald J., Orton, Daniel J., Chrisler, William B., Gaffrey, Matthew J., Liu, Tao, Smith, Richard D., Kelly, Ryan T., Pasa-Tolic, Ljiljana, and Zhu, Ying. Automated Coupling of Nanodroplet Sample Preparation with Liquid Chromatography–Mass Spectrometry for High-Throughput Single-Cell Proteomics. United States: N. p., 2020. Web. doi:10.1021/acs.analchem.0c01551.
Williams, Sarah M., Liyu, Andrey V., Tsai, Chia-Feng, Moore, Ronald J., Orton, Daniel J., Chrisler, William B., Gaffrey, Matthew J., Liu, Tao, Smith, Richard D., Kelly, Ryan T., Pasa-Tolic, Ljiljana, & Zhu, Ying. Automated Coupling of Nanodroplet Sample Preparation with Liquid Chromatography–Mass Spectrometry for High-Throughput Single-Cell Proteomics. United States. https://doi.org/10.1021/acs.analchem.0c01551
Williams, Sarah M., Liyu, Andrey V., Tsai, Chia-Feng, Moore, Ronald J., Orton, Daniel J., Chrisler, William B., Gaffrey, Matthew J., Liu, Tao, Smith, Richard D., Kelly, Ryan T., Pasa-Tolic, Ljiljana, and Zhu, Ying. Wed . "Automated Coupling of Nanodroplet Sample Preparation with Liquid Chromatography–Mass Spectrometry for High-Throughput Single-Cell Proteomics". United States. https://doi.org/10.1021/acs.analchem.0c01551. https://www.osti.gov/servlets/purl/1673582.
@article{osti_1673582,
title = {Automated Coupling of Nanodroplet Sample Preparation with Liquid Chromatography–Mass Spectrometry for High-Throughput Single-Cell Proteomics},
author = {Williams, Sarah M. and Liyu, Andrey V. and Tsai, Chia-Feng and Moore, Ronald J. and Orton, Daniel J. and Chrisler, William B. and Gaffrey, Matthew J. and Liu, Tao and Smith, Richard D. and Kelly, Ryan T. and Pasa-Tolic, Ljiljana and Zhu, Ying},
abstractNote = {Single-cell proteomics can provide critical biological insight into the cellular heterogeneity that is masked by bulk-scale analysis. Here, we have developed a nanoPOTS (nanodroplet processing in one pot for trace samples) platform and demonstrated its broad applicability for single-cell proteomics. However, because of nanoliter-scale sample volumes, the nanoPOTS platform is not compatible with automated LC-MS systems, which significantly limits sample throughput and robustness. To address this challenge, we have developed a nanoPOTS autosampler allowing fully automated sample injection from nanowells to LC-MS systems. We also developed a sample drying, extraction, and loading workflow to enable reproducible and reliable sample injection. The sequential analysis of 20 samples containing 10 ng tryptic peptides demonstrated high reproducibility with correlation coefficients of >0.995 between any two samples. The nanoPOTS autosampler can provide analysis throughput of 9.6, 16, and 24 single cells per day using 120, 60, and 30 min LC gradients, respectively. As a demonstration for single-cell proteomics, the autosampler was first applied to profiling protein expression in single MCF10A cells using a label-free approach. At a throughput of 24 single cells per day, an average of 256 proteins was identified from each cell and the number was increased to 731 when the Match Between Runs algorithm of MaxQuant was used. Using a multiplexed isobaric labeling approach (TMT-11plex), ~77 single cells could be analyzed per day. We analyzed 152 cells from three acute myeloid leukemia cell lines, resulting in a total of 2558 identified proteins with 1465 proteins quantifiable (70% valid values) across the 152 cells. These data showed quantitative single-cell proteomics can cluster cells to distinct groups and reveal functionally distinct differences.},
doi = {10.1021/acs.analchem.0c01551},
journal = {Analytical Chemistry},
number = 15,
volume = 92,
place = {United States},
year = {Wed Jul 08 00:00:00 EDT 2020},
month = {Wed Jul 08 00:00:00 EDT 2020}
}

Works referenced in this record:

The Technology and Biology of Single-Cell RNA Sequencing
journal, May 2015


Single-Cell Transcriptional Analysis
journal, June 2017


Integrated Genomic and Proteomic Analyses of Gene Expression in Mammalian Cells
journal, October 2004

  • Tian, Qiang; Stepaniants, Serguei B.; Mao, Mao
  • Molecular & Cellular Proteomics, Vol. 3, Issue 10
  • DOI: 10.1074/mcp.M400055-MCP200

Clinical potential of mass spectrometry-based proteogenomics
journal, November 2018

  • Zhang, Bing; Whiteaker, Jeffrey R.; Hoofnagle, Andrew N.
  • Nature Reviews Clinical Oncology, Vol. 16, Issue 4
  • DOI: 10.1038/s41571-018-0135-7

On the Dependency of Cellular Protein Levels on mRNA Abundance
journal, April 2016


Integrated Proteome Analysis Device for Fast Single-Cell Protein Profiling
journal, October 2018


SCoPE-MS: mass spectrometry of single mammalian cells quantifies proteome heterogeneity during cell differentiation
journal, October 2018


High-Throughput Single Cell Proteomics Enabled by Multiplex Isobaric Labeling in a Nanodroplet Sample Preparation Platform
journal, September 2019


New mass spectrometry technologies contributing towards comprehensive and high throughput omics analyses of single cells
journal, January 2019

  • Couvillion, Sneha P.; Zhu, Ying; Nagy, Gabe
  • The Analyst, Vol. 144, Issue 3
  • DOI: 10.1039/C8AN01574K

Single-Cell Mass Spectrometry for Discovery Proteomics: Quantifying Translational Cell Heterogeneity in the 16-Cell Frog ( Xenopus ) Embryo
journal, January 2016

  • Lombard-Banek, Camille; Moody, Sally A.; Nemes, Peter
  • Angewandte Chemie International Edition, Vol. 55, Issue 7
  • DOI: 10.1002/anie.201510411

Single Cell Proteomics Using Frog ( Xenopus laevis ) Blastomeres Isolated from Early Stage Embryos, Which Form a Geometric Progression in Protein Content
journal, June 2016


An Improved Boosting to Amplify Signal with Isobaric Labeling (iBASIL) Strategy for Precise Quantitative Single-cell Proteomics
journal, May 2020

  • Tsai, Chia-Feng; Zhao, Rui; Williams, Sarah M.
  • Molecular & Cellular Proteomics, Vol. 19, Issue 5
  • DOI: 10.1074/mcp.RA119.001857

Nanoliter-Scale Oil-Air-Droplet Chip-Based Single Cell Proteomic Analysis
journal, March 2018


Nanodroplet processing platform for deep and quantitative proteome profiling of 10–100 mammalian cells
journal, February 2018


Benchmarking common quantification strategies for large-scale phosphoproteomics
journal, March 2018

  • Hogrebe, Alexander; von Stechow, Louise; Bekker-Jensen, Dorte B.
  • Nature Communications, Vol. 9, Issue 1
  • DOI: 10.1038/s41467-018-03309-6

Nanoproteomics comes of age
journal, October 2018


Improved Single-Cell Proteome Coverage Using Narrow-Bore Packed NanoLC Columns and Ultrasensitive Mass Spectrometry
journal, January 2020


Proteomic Analysis of Single Mammalian Cells Enabled by Microfluidic Nanodroplet Sample Preparation and Ultrasensitive NanoLC-MS
journal, June 2018

  • Zhu, Ying; Clair, Geremy; Chrisler, William B.
  • Angewandte Chemie International Edition, Vol. 57, Issue 38
  • DOI: 10.1002/anie.201802843

Automated mass spectrometry imaging of over 2000 proteins from tissue sections at 100-μm spatial resolution
journal, January 2020


Spatially Resolved Proteome Mapping of Laser Capture Microdissected Tissue with Automated Sample Transfer to Nanodroplets
journal, June 2018

  • Zhu, Ying; Dou, Maowei; Piehowski, Paul D.
  • Molecular & Cellular Proteomics, Vol. 17, Issue 9
  • DOI: 10.1074/mcp.TIR118.000686

The MaxQuant computational platform for mass spectrometry-based shotgun proteomics
journal, October 2016


Fully Automated Four-Column Capillary LC−MS System for Maximizing Throughput in Proteomic Analyses
journal, January 2008

  • Livesay, Eric A.; Tang, Keqi; Taylor, Beverley K.
  • Analytical Chemistry, Vol. 80, Issue 1
  • DOI: 10.1021/ac701727r

Nanowell-mediated multidimensional separations combining nanoLC with SLIM IM-MS for rapid, high-peak-capacity proteomic analyses
journal, November 2018

  • Dou, Maowei; Chouinard, Christopher D.; Zhu, Ying
  • Analytical and Bioanalytical Chemistry, Vol. 411, Issue 21
  • DOI: 10.1007/s00216-018-1452-5

Surface Passivation for Single-molecule Protein Studies
journal, January 2014

  • Chandradoss, Stanley D.; Haagsma, Anna C.; Lee, Young Kwang
  • Journal of Visualized Experiments, Issue 86
  • DOI: 10.3791/50549

Improved Sensitivity in Low-Input Proteomics Using Micropillar Array-Based Chromatography
journal, October 2019


Online Parallel Accumulation–Serial Fragmentation (PASEF) with a Novel Trapped Ion Mobility Mass Spectrometer
journal, November 2018

  • Meier, Florian; Brunner, Andreas-David; Koch, Scarlet
  • Molecular & Cellular Proteomics, Vol. 17, Issue 12
  • DOI: 10.1074/mcp.TIR118.000900