scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Quantitative proteomics: challenges and opportunities in basic and applied research.

TL;DR: The current and future roles of quantitative proteomics in molecular systems biology, clinical research and personalized medicine are explored and existing challenges and limitations are reflected on.
Abstract: In this Perspective, we discuss developments in mass-spectrometry-based proteomic technology over the past decade from the viewpoint of our laboratory We also reflect on existing challenges and limitations, and explore the current and future roles of quantitative proteomics in molecular systems biology, clinical research and personalized medicine

Summary (1 min read)

Jump to: [Introduction] and [Conclusion]

Introduction

  • Proteins constitute a large part of the molecular machinery of the cell and are the major class of biomolecules targeted by drugs.
  • Identified peptide sequences can then be mapped to proteins and the signal intensities of either peptides or fragment ions can be used to estimate relative abundance changes across samples.
  • In 2012, their lab described a new DIA-based method termed SWATH-MS, which uses a targeted paradigm for the analysis of DIA data40.
  • Chemical cross-linking can be used to gain insights into the topology of a protein complex 66-68.
  • To conduct studies at larger scale, proteomic techniques that allow higher throughput, while maintaining robustness, repeatability and sensitivity are therefore essential.

Conclusion

  • Over the past two decades, the authors have witnessed rapid developments in mass spectrometric instrumentation as well as acquisition methods and analysis strategies.
  • Furthermore, quantitative proteomics has contributed enormously to biological and clinically oriented research.
  • Current instrument operation as well as data acquisition and analysis still require highly specialized expertise.
  • Many facilities, including ours, are therefore working towards the development of more robust MS-based methods and automated analysis pipelines to make quantitative proteomics available, not just to expert labs, but also to general molecular biology laboratories in academia, hospitals and industry.

Did you find this useful? Give us your feedback

Figures (3)

Content maybe subject to copyright    Report

Quantitative proteomics: challenges and opportunities in basic and
applied research
Schubert, O. T., Röst, H. L., Collins, B. C., Rosenberger, G., & Aebersold, R. (2017). Quantitative proteomics:
challenges and opportunities in basic and applied research.
Nature Protocols
,
12
(7), 1289-1294.
https://doi.org/10.1038/nprot.2017.040
Published in:
Nature Protocols
Document Version:
Peer reviewed version
Queen's University Belfast - Research Portal:
Link to publication record in Queen's University Belfast Research Portal
Publisher rights
Copyright 2017 Nature Research. This work is made available online in accordance with the publisher’s policies. Please refer to any
applicable terms of use of the publisher.
General rights
Copyright for the publications made accessible via the Queen's University Belfast Research Portal is retained by the author(s) and / or other
copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated
with these rights.
Take down policy
The Research Portal is Queen's institutional repository that provides access to Queen's research output. Every effort has been made to
ensure that content in the Research Portal does not infringe any person's rights, or applicable UK laws. If you discover content in the
Research Portal that you believe breaches copyright or violates any law, please contact openaccess@qub.ac.uk.
Download date:09. Aug. 2022

1
Quantitative proteomics: Challenges
and opportunities in basic and applied research
Olga T. Schubert
1
, Hannes L. Röst
2,5
, Ben C. Collins
3,5
, George Rosenberger
3,5,
Ruedi Aebersold
3,4,
*
1 Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
2 Department of Genetics, Stanford University, Stanford, CA 94305, USA
3 Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
4 Faculty of Science, University of Zurich, 8057 Zurich, Switzerland
5 Authors contributed equally
*Corresponding author (aebersold@imsb.biol.ethz.ch)
Summary
In this perspective, we discuss developments in mass spectrometry-based proteomic
technology in the last decade from the viewpoint of our laboratory. We also reflect on existing
challenges and limitations, and explore the current and future role of quantitative proteomics in
molecular systems biology, clinical research and personalized medicine.

2
Introduction
Proteins constitute a large part of the molecular machinery of the cell and are the major class of
biomolecules targeted by drugs. Organized in functional modules and networks, they carry out
cellular functions and determine phenotypes by means of coordinated activities of a multitude of
molecular species
1
. Traditional biochemical methods for studying proteins have been highly
biased towards a relatively small subset of proteins for which high quality, mainly antibody-
based assays have been available
2
. Over the past two decades, mass spectrometry (MS)-
based methods have emerged as the method of choice for the confident and near exhaustive
identification and quantification of the proteins contained in a biological sample and have
significantly contributed to unraveling cellular signaling networks, to elucidating the dynamics of
protein-protein interactions in different cellular states, and to improved diagnosis and molecular
understanding of disease mechanisms. Overall, MS-based proteomics can reveal the
quantitative state of a proteome and thereby provides insights into the biochemical state of the
respective cell or tissue. In the following paragraphs, we will discuss important concepts and
developments in proteomic technology and explore the current and future role of quantitative
proteomics in molecular systems biology as well as clinical research and personalized medicine.
Evolution of MS-based quantitative proteomics
MS-based proteomics can be broadly grouped into top-down proteomics where intact proteins
are measured and bottom-up proteomics where peptides are measured as surrogates for the
respective protein; in this commentary, we will focus on bottom-up proteomics. The typical
bottom-up proteomics workflow starts with trypsin digestion of a protein sample into short
peptides which are then separated by liquid chromatography either directly or after biochemical
fractionation (Figure 1A)
3
. As peptides elute from the chromatography column, they are
subjected to electrospray ionization
4,5
and are directly sprayed into the mass spectrometer,
where two levels of MS measurement take place in tandem
3
. At the first level, a mass analyzer
measures the mass-to-charge ratio (m/z) of peptide molecular ions (MS1). At the second level,
m/z values of fragment ions resulting from the fragmentation of specific peptide ions are
detected (MS2). The specific fragment ion pattern of each peptide ion together with its m/z value
enable confident identification of peptides present in the sample. Identified peptide sequences
can then be mapped to proteins and the signal intensities of either peptides or fragment ions
can be used to estimate relative abundance changes across samples.

3
Figure 1. Standard MS-based proteomics workflow and acquisition schemes. (A) Proteins can be
extracted from various biological samples, such as bacterial or mammalian cell culture, tissues or bodily
fluids. They are then enzymatically digested into peptides, which are then subjected to reverse-phase
liquid chromatography, ionized with electrospray ionization and sprayed into the mass spectrometer. The
time dimension in B and C is this chromatographic time. (B) Different acquisition schemes for tandem MS
sample the proteome in distinct ways. While the most widely used untargeted (also referred to as shotgun
or data-dependent acquisition, DDA) is relatively simple and applicable to any sample without requiring
prior knowledge, resulting data can suffer from missing data points due to the stochastic sampling
process. In contrast, targeted acquisition acquires peptide and fragment ion data in a highly consistent
manner allowing accurate and sensitive quantification, but is limited to a relatively small, pre-defined set
of peptides. Data-independent acquisition (DIA) acquires data of all detectable fragment ions in a sample
in a systematic and consistent manner, but due to the relatively large peptide ion isolation windows (m/z
dimension) the resulting data is more complex than for the other two acquisition schemes. (C) DIA data
can be analyzed in different ways, either directly analyzing the multiplexed MS2 spectra or first extracting
a subset of informative fragment ion signals (requires prior knowledge) and using these to derive
quantitative data for specific peptides
36,40
.
Discovery-driven
Data-dependent
Hypothesis-driven
Targeted
Data-independent
Time
m/z
B
Time
m/z
C
Time
Intensity
MS2
MS2
MS2
MS2
MS2
Time
Quantitative
MS2 signal
A
m/z
Liquid
chromatography
Peptides Proteins
Tandem mass
spectrometry
Time
Eluent
+
Biological or
clinical samples
MS2
MS1

4
To account for technical variability at various stages of sample handling and during the actual
measurement, in the mid-90s, we and others started to develop strategies based on isotopic
labeling
6,7
, including chemical isotopic labeling
8
, metabolic isotopic labeling
9
, and isobaric
tagging
10,11
. Another important application of isotopic labeling in MS is the use of labeled spike-
in peptides or proteins of known concentration that enable the determination of absolute
concentrations of proteins in a sample, for example, in terms of number of molecules per cell or
nanograms per milliliter of blood
12
. While label-based approaches are still the gold-standard for
quantification by MS-based proteomic methods
13
, the past years have seen label-free
approaches becoming more popular thanks to simpler experimental design and sample
preparation
6,14
. Among the developments enabling this transition are the advance of
commercially available high resolution and fast scanning instruments, such as the introduction
of the Orbitrap (2005)
15
and continuous improvement of time-of-flight mass spectrometers
16
,
combined with improvements in software for aligning multiple MS runs
17,18
. Another more recent
trend, starting in 2006
19
, is label-free absolute quantification, where the absolute concentrations
of all proteins measured in a sample are estimated based on summarized ion counts, which can
then be converted into a meaningful unit by comparison to the total amount of protein that was
injected into the mass spectrometer or by correlation to a set of spiked-in reference peptides of
known concentration
20-23
.
Regardless of whether label-based or label-free strategies are used, bottom-up proteomic
methods have traditionally been divided into discovery proteomics and targeted proteomics
(Figure 1B). Discovery proteomics (also known as shotgun proteomics and exemplified by data-
dependent acquisition, DDA) has its strength in identifying thousands of proteins per run.
However, in complex samples, we have often been faced by limitations regarding repeatability
of peptide identification and consistency of quantification
24,25
. Recent developments in
chromatographic performance and MS hardware alleviate some of these concerns and allow
high-quality quantitative measurements of near-complete proteomes, even in highly complex
samples such as human cell lines and tissues
26-29
.
About a decade ago, in order to overcome the limited scalability and reproducibility of discovery
proteomics in studies aiming to quantify proteins in cohorts consisting of large numbers of
samples, we and others started exploring the capabilities of targeted proteomics (exemplified by
selected/multiple reaction monitoring, S/MRM
30,31
, and more recently parallel reaction
monitoring, PRM
32,33
). Targeting methods provide consistent and accurate quantification, even

Citations
More filters
Journal ArticleDOI
TL;DR: This protocol describes pathway enrichment analysis of gene lists from RNA-seq and other genomics experiments using g:Profiler, GSEA, Cytoscape and EnrichmentMap software, and describes innovative visualization techniques.
Abstract: Pathway enrichment analysis helps researchers gain mechanistic insight into gene lists generated from genome-scale (omics) experiments. This method identifies biological pathways that are enriched in a gene list more than would be expected by chance. We explain the procedures of pathway enrichment analysis and present a practical step-by-step guide to help interpret gene lists resulting from RNA-seq and genome-sequencing experiments. The protocol comprises three major steps: definition of a gene list from omics data, determination of statistically enriched pathways, and visualization and interpretation of the results. We describe how to use this protocol with published examples of differentially expressed genes and mutated cancer genes; however, the principles can be applied to diverse types of omics data. The protocol describes innovative visualization techniques, provides comprehensive background and troubleshooting guidelines, and uses freely available and frequently updated software, including g:Profiler, Gene Set Enrichment Analysis (GSEA), Cytoscape and EnrichmentMap. The complete protocol can be performed in ~4.5 h and is designed for use by biologists with no prior bioinformatics training.

958 citations

Journal ArticleDOI
TL;DR: This study demonstrates that micro-flow LC–MS/MS is suitable for a broad range of proteomic applications and enables robust and reproducible high-throughput proteomics experiments at a very moderate loss of sensitivity.
Abstract: Nano-flow liquid chromatography tandem mass spectrometry (nano-flow LC-MS/MS) is the mainstay in proteome research because of its excellent sensitivity but often comes at the expense of robustness. Here we show that micro-flow LC-MS/MS using a 1 × 150 mm column shows excellent reproducibility of chromatographic retention time ( 2000 samples of human cell lines, tissues and body fluids. Deep proteome analysis identifies >9000 proteins and >120,000 peptides in 16 h and sample multiplexing using tandem mass tags increases throughput to 11 proteomes in 16 h. The system identifies >30,000 phosphopeptides in 12 h and protein-protein or protein-drug interaction experiments can be analyzed in 20 min per sample. We show that the same column can be used to analyze >7500 samples without apparent loss of performance. This study demonstrates that micro-flow LC-MS/MS is suitable for a broad range of proteomic applications.

202 citations

Journal ArticleDOI
TL;DR: The wild-type aerolysin nanopore enables the size-discrimination of several short uniformly charged homopeptides, mixed in solution, with a single amino acid resolution, which is very sensitive, allowing detecting and characterizing a few dozens of peptide impurities in a high purity commercial peptide sample, while conventional analysis techniques fail to do so.
Abstract: There are still unmet needs in finding new technologies for biomedical diagnostic and industrial applications. A technology allowing the analysis of size and sequence of short peptide molecules of only few molecular copies is still challenging. The fast, low-cost and label-free single-molecule nanopore technology could be an alternative for addressing these critical issues. Here, we demonstrate that the wild-type aerolysin nanopore enables the size-discrimination of several short uniformly charged homopeptides, mixed in solution, with a single amino acid resolution. Our system is very sensitive, allowing detecting and characterizing a few dozens of peptide impurities in a high purity commercial peptide sample, while conventional analysis techniques fail to do so.

185 citations

Journal ArticleDOI
TL;DR: In this article, the authors describe the techniques and sources of omics data, outline network theory, and highlight exemplars of novel approaches that combine gene regulatory and co-expression networks, proteomics, metabolomics, lipidomics and phenomics with informatics techniques to provide new insights into cardiovascular disease.
Abstract: Omics techniques generate large, multidimensional data that are amenable to analysis by new informatics approaches alongside conventional statistical methods. Systems theories, including network analysis and machine learning, are well placed for analysing these data but must be applied with an understanding of the relevant biological and computational theories. Through applying these techniques to omics data, systems biology addresses the problems posed by the complex organization of biological processes. In this Review, we describe the techniques and sources of omics data, outline network theory, and highlight exemplars of novel approaches that combine gene regulatory and co-expression networks, proteomics, metabolomics, lipidomics and phenomics with informatics techniques to provide new insights into cardiovascular disease. The use of systems approaches will become necessary to integrate data from more than one omic technique. Although understanding the interactions between different omics data requires increasingly complex concepts and methods, we argue that hypothesis-driven investigations and independent validation must still accompany these novel systems biology approaches to realize their full potential.

102 citations

Journal ArticleDOI
TL;DR: This work implemented single‐pot solid‐phase‐enhanced sample preparation on a liquid handling robot for automated processing (autoSP3) of tissue lysates in a 96‐well format, enabling reproducible tissue proteomics in a broad range of clinical and non‐clinical applications.
Abstract: High-throughput and streamlined workflows are essential in clinical proteomics for standardized processing of samples from a variety of sources, including fresh-frozen tissue, FFPE tissue, or blood. To reach this goal, we have implemented single-pot solid-phase-enhanced sample preparation (SP3) on a liquid handling robot for automated processing (autoSP3) of tissue lysates in a 96-well format. AutoSP3 performs unbiased protein purification and digestion, and delivers peptides that can be directly analyzed by LCMS, thereby significantly reducing hands-on time, reducing variability in protein quantification, and improving longitudinal reproducibility. We demonstrate the distinguishing ability of autoSP3 to process low-input samples, reproducibly quantifying 500-1,000 proteins from 100 to 1,000 cells. Furthermore, we applied this approach to a cohort of clinical FFPE pulmonary adenocarcinoma (ADC) samples and recapitulated their separation into known histological growth patterns. Finally, we integrated autoSP3 with AFA ultrasonication for the automated end-to-end sample preparation and LCMS analysis of 96 intact tissue samples. Collectively, this constitutes a generic, scalable, and cost-effective workflow with minimal manual intervention, enabling reproducible tissue proteomics in a broad range of clinical and non-clinical applications.

93 citations


Cites background from "Quantitative proteomics: challenges..."

  • ...Collectively, this has propelled proteomic studies in multiple areas of basic and mechanistic biology, using deep and quantitative proteomic profiles to understand spatial and temporal aspects of proteome organization and dynamics in a wide variety of conditions (Schubert et al, 2017)....

    [...]

References
More filters
Journal ArticleDOI
TL;DR: MaxQuant, an integrated suite of algorithms specifically developed for high-resolution, quantitative MS data, detects peaks, isotope clusters and stable amino acid isotope–labeled (SILAC) peptide pairs as three-dimensional objects in m/z, elution time and signal intensity space and achieves mass accuracy in the p.p.b. range.
Abstract: Efficient analysis of very large amounts of raw data for peptide identification and protein quantification is a principal challenge in mass spectrometry (MS)-based proteomics. Here we describe MaxQuant, an integrated suite of algorithms specifically developed for high-resolution, quantitative MS data. Using correlation analysis and graph theory, MaxQuant detects peaks, isotope clusters and stable amino acid isotope-labeled (SILAC) peptide pairs as three-dimensional objects in m/z, elution time and signal intensity space. By integrating multiple mass measurements and correcting for linear and nonlinear mass offsets, we achieve mass accuracy in the p.p.b. range, a sixfold increase over standard techniques. We increase the proportion of identified fragmentation spectra to 73% for SILAC peptide pairs via unambiguous assignment of isotope and missed-cleavage state and individual mass precision. MaxQuant automatically quantifies several hundred thousand peptides per SILAC-proteome experiment and allows statistically robust identification and quantification of >4,000 proteins in mammalian cell lysates.

12,340 citations

Journal ArticleDOI
06 Oct 1989-Science
TL;DR: Spectra have been obtained for biopolymers including oligonucleotides and proteins, the latter having molecular weights up to 130,000, with as yet no evidence of an upper limit.
Abstract: Electrospray ionization has recently emerged as a powerful technique for producing intact ions in vacuo from large and complex species in solution. To an extent greater than has previously been possible with the more familiar "soft" ionization methods, this technique makes the power and elegance of mass spectrometric analysis applicable to the large and fragile polar molecules that play such vital roles in biological systems. The distinguishing features of electrospray spectra for large molecules are coherent sequences of peaks whose component ions are multiply charged, the ions of each peak differing by one charge from those of adjacent neighbors in the sequence. Spectra have been obtained for biopolymers including oligonucleotides and proteins, the latter having molecular weights up to 130,000, with as yet no evidence of an upper limit.

6,765 citations

Journal ArticleDOI
13 Mar 2003-Nature
TL;DR: The ability of mass spectrometry to identify and, increasingly, to precisely quantify thousands of proteins from complex samples can be expected to impact broadly on biology and medicine.
Abstract: Recent successes illustrate the role of mass spectrometry-based proteomics as an indispensable tool for molecular and cellular biology and for the emerging field of systems biology. These include the study of protein-protein interactions via affinity-based isolations on a small and proteome-wide scale, the mapping of numerous organelles, the concurrent description of the malaria parasite genome and proteome, and the generation of quantitative protein profiles from diverse species. The ability of mass spectrometry to identify and, increasingly, to precisely quantify thousands of proteins from complex samples can be expected to impact broadly on biology and medicine.

6,597 citations

Journal ArticleDOI
TL;DR: SILAC is a simple, inexpensive, and accurate procedure that can be used as a quantitative proteomic approach in any cell culture system and is applied to the relative quantitation of changes in protein expression during the process of muscle cell differentiation.

5,653 citations

Journal ArticleDOI
TL;DR: It is found that inactivation of Upf1p and Xrn1p causes common as well as unique effects on protein expression, and the use of 4-fold multiplexing to enable relative protein measurements simultaneously with determination of absolute levels of a target protein using synthetic isobaric peptide standards.

4,411 citations

Frequently Asked Questions (2)
Q1. What contributions have the authors mentioned in the paper "Quantitative proteomics: challenges and opportunities in basic and applied research" ?

For instance, mass spectrometer-based methods have emerged as the method of choice for the confident and near exhaustive identification and quantification of the proteins contained in a biological sample and have significantly contributed to unraveling cellular signaling networks, to elucidating the dynamics of proteins-protein interactions in different cellular states, and to improved diagnosis and molecular understanding of disease mechanisms this paper. 

The authors also reflect on existingchallenges and limitations, and explore the current and future role of quantitative proteomics inmolecular systems biology, clinical research and personalized medicine.