Your browser doesn't support javascript.
loading
Optimized data-independent acquisition approach for proteomic analysis at single-cell level.
Wang, Yuefan; Lih, Tung-Shing Mamie; Chen, Lijun; Xu, Yuanwei; Kuczler, Morgan D; Cao, Liwei; Pienta, Kenneth J; Amend, Sarah R; Zhang, Hui.
Affiliation
  • Wang Y; Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA.
  • Lih TM; Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA.
  • Chen L; Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA.
  • Xu Y; Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA.
  • Kuczler MD; Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA.
  • Cao L; Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA.
  • Pienta KJ; Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA.
  • Amend SR; Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, 600 N. Wolfe Street, Baltimore, MD, 21287, USA.
  • Zhang H; Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA. huizhang@jhu.edu.
Clin Proteomics ; 19(1): 24, 2022 Jul 09.
Article in En | MEDLINE | ID: mdl-35810282
ABSTRACT

BACKGROUND:

Single-cell proteomic analysis provides valuable insights into cellular heterogeneity allowing the characterization of the cellular microenvironment which is difficult to accomplish in bulk proteomic analysis. Currently, single-cell proteomic studies utilize data-dependent acquisition (DDA) mass spectrometry (MS) coupled with a TMT labelled carrier channel. Due to the extremely imbalanced MS signals among the carrier channel and other TMT reporter ions, the quantification is compromised. Thus, data-independent acquisition (DIA)-MS should be considered as an alternative approach towards single-cell proteomic study since it generates reproducible quantitative data. However, there are limited reports on the optimal workflow for DIA-MS-based single-cell analysis.

METHODS:

We report an optimized DIA workflow for single-cell proteomics using Orbitrap Lumos Tribrid instrument. We utilized a breast cancer cell line (MDA-MB-231) and induced drug resistant polyaneuploid cancer cells (PACCs) to evaluate our established workflow.

RESULTS:

We found that a short LC gradient was preferable for peptides extracted from single cell level with less than 2 ng sample amount. The total number of co-searching peptide precursors was also critical for protein and peptide identifications at nano- and sub-nano-gram levels. Post-translationally modified peptides could be identified from a nano-gram level of peptides. Using the optimized workflow, up to 1500 protein groups were identified from a single PACC corresponding to 0.2 ng of peptides. Furthermore, about 200 peptides with phosphorylation, acetylation, and ubiquitination were identified from global DIA analysis of 100 cisplatin resistant PACCs (20 ng). Finally, we used this optimized DIA approach to compare the whole proteome of MDA-MB-231 parental cells and induced PACCs at a single-cell level. We found the single-cell level comparison could reflect real protein expression changes and identify the protein copy number.

CONCLUSIONS:

Our results demonstrate that the optimized DIA pipeline can serve as a reliable quantitative tool for single-cell as well as sub-nano-gram proteomic analysis.

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Clin Proteomics Year: 2022 Type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Clin Proteomics Year: 2022 Type: Article Affiliation country: United States