Elsevier

Journal of Chromatography A

Volume 1523, 10 November 2017, Pages 40-48
Journal of Chromatography A

Review article
Advances in microscale separations towards nanoproteomics applications

https://doi.org/10.1016/j.chroma.2017.07.055Get rights and content

Highlights

  • Nanoproteomics represents a daunting analytical challenge in terms of sensitivity.

  • NanoLC and CE coupled with MS can provide single cell level sensitivity.

  • Front-end sample processing is the current bottleneck of overall sensitivity in MS proteomics.

  • Significant advances have achieved towards nanoproteomics measurements.

  • Further advances are still required for enabling broad nanoproteomics applications.

Abstract

Microscale separation (e.g., liquid chromatography or capillary electrophoresis) coupled with mass spectrometry (MS) has become the primary tool for advanced proteomics, an indispensable technology for gaining understanding of complex biological processes. In recent decades significant advances have been achieved in MS-based proteomics. However, the current proteomics platforms still face an analytical challenge in overall sensitivity towards nanoproteomics applications for starting materials of less than 1 μg total proteins (e.g., cellular heterogeneity in tissue pathologies). Herein, we review recent advances in microscale separation techniques and integrated sample processing strategies that improve the overall sensitivity and proteome coverage of the proteomics workflow, and their contributions towards nanoproteomics applications.

Introduction

Proteins are the workhorses of the cell, impacting nearly all aspects of cellular processes. The proteome by its nature is dynamic, and the acute state of the proteome (i.e., the proteotype) depends on both the genotype and external perturbations [1], [2]. Therefore, quantitative analysis of the dynamics of the proteome including post-translational modifications (PTMs) and its connection to phenotypes (e.g., diseases) has become indispensable in biological and clinical research [3], [4], [5]. Recent advances in mass spectrometry (MS)-based proteomics for both global deep-profiling of the proteome and selected types of PTMs (e.g., phosphorylation) [6], [7], [8] and targeted quantification of proteins from specific signaling pathways [9], [10] have greatly expanded our capabilities in performing proteogenomics and systems biology studies for gaining detailed mechanistic insights into physiological and pathological processes.

During the past decade, major advances have been achieved in nearly all areas of the proteomics workflow such as high resolution microscale chromatographic separations, mass spectrometry instrumentation, and bioinformatics data analysis tools to enable large-scale proteome interrogation [11]. Current state-of-the-art MS-based proteomics platforms can afford deep coverage for both the global proteome and selected PTMs in cell or tissue samples. For example, recent studies have reported the identification or quantification of ∼10,000 proteins [6], [7], >20,000 phosphorylation [12], [13] and >15,000 ubiquitination sites [12], [14].

Despite recent advances in improving overall proteome coverage, the current proteomics workflows typically require relatively large amounts of starting materials on the order of millions of cells or 100 s μg of proteins, which excludes many important biological and biomedical applications. The ability to effectively analyze extremely small amounts of protein samples (e.g., nanograms of proteins) from cells or tissues by MS is one of the most significant challenges for current MS-proteomics. Herein, we define sample amounts with less than 1 μg of total protein as “nanoscale” and proteomics analyses of these nanoscale samples as “nanoproteomics” (Fig. 1). The biomedical need for nanoproteomics technologies are compelling, including the analyses of tissue substructures, cellular microenvironments of disease pathologies, rare or small subpopulations of cells, extracellular vesicles, as well as single cell resolution profiling (Fig. 1). Some of these sample types are readily produced by existing technologies such as fluorescence activated cell sorting (FACS) [15], laser capture dissection (LCM) [16], [17], and exosome isolation techniques [18]. Moreover, single cell resolution genomics technologies, such as single-cell genomic sequencing [19] and single-cell transcriptomic profiling (RNA-Seq) [20], [21], have been making tremendous impact in biological research. However, the current state-of-the-art in MS-based proteomics still falls far short of the sensitivity required for single cell analyses.

Considerable efforts have been devoted to enhance the overall sensitivity of MS-based proteomics workflow towards enabling analysis of small samples, including the front-end sample processing, microscale separations, and MS instrumentation. Herein, we review recent advances in microscale separation, as well as nanoscale sample processing systems for proteomics analysis. Our focus will be on bottom-up proteomics, and the other important advances in top-down proteomics (measurement of intact proteins) [22] are not covered here.

Section snippets

Factors governing overall MS-based proteomics sensitivity

Fig. 2 illustrates a typical MS-based proteomics workflow for protein identification in biological samples. Conceptually, the overall analytical sensitivity of MS-based proteomics depends on the following aspects: a) the efficiency and recovery of front-end sample processing (e.g., protein extraction and protein digestion) and the degree of reducing sample complexity by extensive fractionation and/or enrichment; b) the resolving power of chromatographic or electrophoretic separations when

LC or CE for nanoproteomics

Reversed phase (RP) LC, either in packed columns or monolithic columns, is the most widely used LC separation for bottom-up proteomics due to its relatively high resolving power and ESI friendly mobile phases. CE has also emerged as a powerful separation technique when directly coupled with ESI-MS for proteomic research. One of the foundational discoveries in ESI-MS was that the ESI efficiency can be dramatically enhanced by nearly ∼100-fold when ESI was operated at low flow rate (20–40 nL/min)

Front-end microscale sample processing

As a critical component in the overall workflow, there has been a significant interest in developing microscale sample processing techniques to minimize sample loss and increase processing efficiency. In principle, this would involve processing with minimized liquid volumes and transfer steps. These efforts generally employ two main approaches to reduce sample losses and enhance processing efficiency: single-tube preparation techniques or integrated online processing systems.

The single tube

Highlights of nanoproteomics applications

Nanoproteomics is an evolving technological capability for enabling analysis of cellular heterogeneity, tissue substructures, and other nanoscale biological or clinical samples at the proteome level when coupled with cell isolation techniques (e.g., LCM [97] and FACS [98]). While we recognized that most of the reports on nanoproteomics were focused on method development or proof-of-concept demonstrations, in this section we highlight some studies to illustrate the potential of biological

Conclusions and perspectives

Tremendous advances in LC- and CE-MS platforms have been achieved in terms of overall sensitivity, proteome coverage, reproducibility, and quantification for global proteome analyses. The absolute sensitivity for LC-MS and CE-MS operating in the nanoflow regime is sufficient for analyzing low ng protein samples or small numbers of cells, and potentially even for single mammalian cells [29], [30]. The main bottleneck for the overall sensitivity lies in the sample losses and efficiency in the

Conflict of interest

None.

Acknowledgements

Portions of this work were supported by the NIH by NIH Grants P41 GM103493, DP3 DK110844, and UC4 DK104167. The experimental work described herein was performed in the Environmental Molecular Sciences Laboratory, a national scientific user facility sponsored by the DOE and located at Pacific Northwest National Laboratory, which is operated by Battelle Memorial Institute for the DOE under Contract DE-AC05-76RL0 1830.

References (113)

  • C. Xie et al.

    Octadecylated silica monolith capillary column with integrated nanoelectrospray ionization emitter for highly efficient proteome analysis

    Mol. Cell. Proteomics

    (2006)
  • G. Zhu et al.

    On-line amino acid-based capillary isoelectric focusing-ESI–MS/MS for protein digests analysis

    Anal. Chim. Acta

    (2012)
  • L. Sun et al.

    Capillary zone electrophoresis-electrospray ionization-tandem mass spectrometry for quantitative parallel reaction monitoring of peptide abundance and single-shot proteomic analysis of a human cell line

    J. Chromatogr. A

    (2014)
  • N.A. Kulak et al.

    Loss-less nano-fractionator for high sensitivity, high coverage proteomics

    Mol. Cell. Proteomics

    (2017)
  • R.B. Braakman et al.

    Optimized nLC-MS workflow for laser capture microdissected breast cancer tissue

    J. Proteomics

    (2012)
  • S. Jiang et al.

    A one-step preparation method of monolithic enzyme reactor for highly efficient sample preparation coupled to mass spectrometry-based proteomics studies

    J. Chromatogr. A

    (2015)
  • M. Safdar et al.

    Microscale immobilized enzyme reactors in proteomics: latest developments

    J. Chromatogr. A

    (2014)
  • L. Yan et al.

    In-tip nanoreactors for cancer cells proteome profiling

    Anal. Chim. Acta

    (2017)
  • R. Aebersold et al.

    Mass-spectrometric exploration of proteome structure and function

    Nature

    (2016)
  • A.F. Altelaar et al.

    Next-generation proteomics: towards an integrative view of proteome dynamics

    Nat. Rev. Genet.

    (2013)
  • M. Larance et al.

    Multidimensional proteomics for cell biology

    Nat. Rev. Mol. Cell Biol.

    (2015)
  • P. Mertins et al.

    Proteogenomics connects somatic mutations to signalling in breast cancer

    Nature

    (2016)
  • B. Zhang et al.

    Proteogenomic characterization of human colon and rectal cancer

    Nature

    (2014)
  • T. Shi et al.

    Conservation of protein abundance patterns reveals the regulatory architecture of the EGFR-MAPK pathway

    Sci. Signal.

    (2016)
  • N. Bisson et al.

    Selected reaction monitoring mass spectrometry reveals the dynamics of signaling through the GRB2 adaptor

    Nat. Biotechnol.

    (2011)
  • P. Mertins et al.

    Integrated proteomic analysis of post-translational modifications by serial enrichment

    Nat. Methods

    (2013)
  • C. Gawad et al.

    Single-cell genome sequencing: current state of the science

    Nat. Rev. Genet.

    (2016)
  • E.S. Drummond et al.

    Proteomic analysis of neurons microdissected from formalin-fixed, paraffin-embedded Alzheimer's disease brain tissue

    Sci. Rep.

    (2015)
  • G. Clair et al.

    Spatially-resolved proteomics: rapid quantitative analysis of laser capture microdissected alveolar tissue samples

    Sci. Rep.

    (2016)
  • M.A. Livshits et al.

    Isolation of exosomes by differential centrifugation: theoretical analysis of a commonly used protocol

    Sci. Rep.

    (2016)
  • E. Shapiro et al.

    Single-cell sequencing-based technologies will revolutionize whole-organism science

    Nat. Rev. Genet.

    (2013)
  • T. Kalisky et al.

    Genomic analysis at the single-cell level

    Annu. Rev. Genet.

    (2011)
  • J.G. Lohr et al.

    Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer

    Nat. Biotechnol.

    (2014)
  • T.K. Toby et al.

    Progress in top-down proteomics and the analysis of proteoforms

    Annu. Rev. Anal. Chem.

    (2016)
  • S.A. Shaffer et al.

    Characterization of a new electrodynamic ion funnel interface for electrospray ionization mass spectrometry

    Anal. Chem.

    (1999)
  • A. Michalski et al.

    Mass spectrometry-based proteomics using Q Exactive, a high-performance benchtop quadrupole Orbitrap mass spectrometer

    Mol. Cell. Proteomics

    (2011)
  • Y. Zhang et al.

    Protein analysis by shotgun/bottom-up proteomics

    Chem. Rev.

    (2013)
  • M. Wilm et al.

    Analytical properties of the nanoelectrospray ion source

    Anal. Chem.

    (1996)
  • Y. Shen et al.

    Ultrasensitive proteomics using high-efficiency on-line micro-SPE-NanoLC-NanoESI MS and MS/MS

    Anal. Chem.

    (2004)
  • L. Sun et al.

    Ultrasensitive and fast bottom-up analysis of femtogram amounts of complex proteome digests

    Angew. Chem. Int. Ed. Engl.

    (2013)
  • Y. Shen et al.

    High-efficiency nanoscale liquid chromatography coupled on-line with mass spectrometry using nanoelectrospray ionization for proteomics

    Anal. Chem.

    (2002)
  • L. Zang et al.

    Proteomic analysis of ductal carcinoma of the breast using laser capture microdissection LC-MS, and 16O/18O isotopic labeling

    J. Proteome Res.

    (2004)
  • L.F. Waanders et al.

    Quantitative proteomic analysis of single pancreatic islets

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • L. Sun et al.

    Single cell proteomics using frog (Xenopus laevis) blastomeres isolated from early stage embryos, which form a geometric progression in protein content

    Anal. Chem.

    (2016)
  • N. Wang et al.

    Development of mass spectrometry-based shotgun method for proteome analysis of 500–5000 cancer cells

    Anal. Chem.

    (2010)
  • A.R. Ivanov et al.

    Low-attomole electrospray ionization MS and MS/MS analysis of protein tryptic digests using 20-μm-i.d. polystyrene-divinylbenzene monolithic capillary columns

    Anal. Chem.

    (2003)
  • Q. Luo et al.

    Preparation of 20 μm-i.d. silica-based monolithic columns and their performance for proteomics analyses

    Anal. Chem.

    (2005)
  • Q. Luo et al.

    MicroSPE-nanoLC-ESI–MS/MS using 10-mu m-i.d. Silica-based monolithic columns for proteomics

    Anal. Chem.

    (2007)
  • Q. Luo et al.

    More sensitive and quantitative proteomic measurements using very low flow rate porous silica monolithic LC columns with electrospray ionization-mass spectrometry

    J. Proteome Res.

    (2006)
  • Q. Luo et al.

    On-line 1D and 2D porous layer open tubular/LC-ESI–MS using 10-μm-i.d. poly(styrene-divinylbenzene) columns for ultrasensitive proteomic analysis

    Anal. Chem.

    (2007)
  • Cited by (41)

    • Nano-liquid chromatography

      2023, Liquid Chromatography: Fundamentals and Instrumentation: Volume 1, Third Edition
    • Liquid chromatography in proteomics research

      2023, Liquid Chromatography: Applications
    • A review on recent trends in the phosphoproteomics workflow. From sample preparation to data analysis

      2022, Analytica Chimica Acta
      Citation Excerpt :

      Also, the significant number of capillary columns with particulate stationary phases presented in this review were self-packed in research labs, confirming that non-commercially packed columns might be successfully applied in both global and targeted proteomics studies. On the other hand, significant efforts are still necessary to improve the quality of the proteomic separations, especially in terms of the phosphorylation post-translational modification analysis in nanoscale and single-cell samples, where the sub-stoichiometric and low-abundance nature of PTMs provides yet another sensitivity challenge [116]. In untargeted proteomics, the main aim is to utilize mass spectrometry to characterize and quantify as many proteins in the complex sample as possible.

    • Capillary ultrahigh-pressure liquid chromatography-mass spectrometry for fast and high resolution metabolomics separations

      2021, Journal of Chromatography A
      Citation Excerpt :

      Capillary LC columns can also provide better MS sensitivity with reduced ion suppression because of lower flow rates [34], utility for sample-limited analysis, and economical use of mobile and stationary phases [35]. Capillary columns are routinely used in proteomics but have not yet been widely implemented in metabolomics [36,37]. Benzoyl chloride (BzCl) derivatization was used to improve retention of polar metabolites on reversed phase columns, as such labeling strategies have shown to be useful for both targeted and untargeted workflows [38–40].

    View all citing articles on Scopus
    View full text