JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY
VOL. 68, NO. 25, 2016
PUBLISHED BY ELSEVIER ON BEHALF OF THE AMERICAN
ISSN 0735-1097/$36.00
COLLEGE OF CARDIOLOGY FOUNDATION
http://dx.doi.org/10.1016/j.jacc.2016.10.031
FOCUS SEMINAR: GENETICS STATE-OF-THE-ART REVIEW
Proteomics Research in Cardiovascular Medicine and Biomarker Discovery Maggie P.Y. Lam, PHD,a Peipei Ping, PHD,a Elizabeth Murphy, PHDb
ABSTRACT Proteomics is a systems physiology discipline to address the large-scale characterization of protein species within a biological system, be it a cell, a tissue, a body biofluid, an organism, or a cohort population. Building on advances from chemical analytical platforms (e.g., mass spectrometry and other technologies), proteomics approaches have contributed powerful applications in cardiovascular biomedicine, most notably in: 1) the discovery of circulating protein biomarkers of heart diseases from plasma samples; and 2) the identification of disease mechanisms and potential therapeutic targets in cardiovascular tissues, in both preclinical models and translational studies. Contemporary proteomics investigations offer powerful means to simultaneously examine tens of thousands of proteins in various samples, and understand their molecular phenotypes in health and disease. This concise review introduces study design considerations, example applications and use cases, as well as interpretation and analysis of proteomics data in cardiovascular biomedicine. (J Am Coll Cardiol 2016;68:2819–30) Published by Elsevier on behalf of the American College of Cardiology Foundation.
O
ver the past 20 years, there has been accel-
molecular changes at the protein layer, in particular,
erating
can provide independent insights into disease mech-
growth
in
the
application
of
proteomics in cardiovascular biomedicine,
anisms in the following 3 areas.
building on pioneering studies with translational
First, it has gradually emerged from large-scale
significance (1,2) and mechanistic insights (3–8).
studies that transcript and protein changes in a
The majority of human genes functions to create
system may, at times, correspond poorly—with
proteins, which are the molecular workhorses that
transcript levels explaining as little as 10% to 30% of
carry out virtually all metabolic, signaling, and phys-
variations in protein abundance. Although the exact
iological functions in life. Many human diseases
contribution of transcripts to protein-level abun-
may be characterized by the elicited changes in
dance is debated, it is evident that in the heart and
proteome configurations. In complex late onset dis-
other organs, a large number of post-transcriptional
eases
environmental
modulators can alter disease-driver protein expres-
components exist, such that individual genetic vari-
sion and function during pathogenesis, without
in
particular,
considerable
ants are often poorly predictive of disease states.
changes
Telltale molecular signatures of acquired cardiovas-
modulators include components of the nonsense-
cular diseases may instead manifest through inter-
mediated decay pathway, long noncoding RNAs,
mediate
and microRNAs (11–14).
phenotypes,
including
transcript
and
protein abundance. Beyond what may be learned from
gene
and
transcript
information
alone,
in
transcript
abundance
(9,10).
These
Second, plasma proteins provide an accessible readout of the status of potentially all tissues, and
Listen to this manuscript’s audio summary by JACC Editor-in-Chief Dr. Valentin Fuster. From the aNIH BD2K Center of Excellence and Department of Physiology, Medicine and Bioinformatics, University of California, Los Angeles, California; and the bSystems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland. This work was supported by NIH K99-HL127302 to Dr. Lam; NIH R37-HL063901 and NIH U54-GM114833 to Dr. Ping; and NIH ZIA-HL006059 and NIH ZIA-HL002066 to Dr. Murphy. Manuscript received August 3, 2016; revised manuscript received October 20, 2016, accepted October 21, 2016.
2820
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
Proteomics in Cardiovascular Research
ABBREVIATIONS
are the sources of many current biomarkers
in plasma may be viewed as classical resident
AND ACRONYMS
in use (Table 1). Clinically useful biomarkers
plasma proteins, which tend to occupy the higher
can
end of the abundance spectrum. These resident
AP-MS = affinity purificationmass spectrometry
FDR = false discovery rate
include
circulating
proteins
that
have been secreted or leaked directly into
proteins
the plasma from resident cells (e.g., myo-
such as albumin, immunoglobulins, transferrins,
include
extremely
abundant
species,
cytes) following diseases or injuries (15), and
and fibrinogens, which are found in the milligram/
are thus spatially uncoupled from the tran-
milliliter concentration range and account for >90%
script change at the tissue of origin. The
of plasma protein content. Other plasma proteins
tissues of origin of circulating proteins may
come from secretion or leakage from various organs
modification
be unknown, inaccessible, or invasive to
before being diluted into the 5 liters of peripheral
RNA-seq = ribonucleic acid-
procure,
hunt
blood, and are found in the microgram/milliliter
sequencing
for these potential biomarkers via trans-
to nanogram/milliliter range. Very low-abundance
TMT = tandem mass tag
cript
proteins, including interleukin-6 and -12, tumor
MS = mass spectrometry PDH = pyruvate dehydrogenase
PTM = post-translational
making
it
impractical
measurements,
instead
to
of
protein
measurements.
necrosis factor-alpha, and other cytokines, circulate
Third, disease processes may be mediated not by
in the picogram/milliliter range, and are typically
the altered abundance of gene products, but rather
masked by higher-abundance proteins in untargeted
by other functional parameters of the proteome,
proteomics analyses (Figure 1). Adding to this
including protein post-translational modifications
complexity, plasma proteins may be proteolytically
(PTMs), protein-protein interactions, and protein
processed, creating numerous degradation product
degradation, which take place after the protein
species, and repetitive sequences in proteins can
molecules are synthesized and cannot be predicted
give a spurious appearance of higher abundance.
from genetic information a priori.
These challenges are compounded by the absence of
Proteomics
to
a protein equivalent of the polymerase chain reac-
measure protein function on a large scale, and hence
tion, such that amplification of minute samples is
interrogate
not possible.
abuts
technologies the
allow
molecular
physiological
layer
that
In the following 3 segments of this review, we will
the complexity of the human proteome also presents
discuss 3 aspects of proteomics applications in car-
a
analytical
challenge
(16).
closely However,
daunting
phenotypes
researchers
large-scale
diovascular biomedical research: 1) study design for
characterization. For example, circulating proteins
for
biomarker research and discovery, with discussions
in human plasma, found at a concentration of
on approaches to circumvent analytical challenges; 2)
w70 mg/ml, are estimated to comprise at least
characterization of multidimensional protein param-
10,000 distinct protein species (17) over a concen-
eters in disease mechanism research; and 3) inter-
tration range of >10 orders of magnitude (10-billion-
pretation and validation of proteomics data or
fold differences) (18). Approximately 3,000 proteins
methodologies.
T A B L E 1 Selected Protein Biomarkers for Cardiovascular Diseases
Protein Biomarker
Abbreviation
Disease Relevance
Required Assay Sensitivity (Estimate)
Discovery Period*
Ref. #
Apolipoprotein A-I
APOA
Cardiovascular event risk
w1 mg/ml
Mid-1980s
(96)
Apolipoprotein B
APOB
Cardiovascular event risk
w1 mg/ml
Mid-1980s
(97)
B-type natriuretic peptide
BNP
Heart failure, acute coronary syndrome
w100 pg/ml
Early 2000s
(98)
C-reactive protein
CRP
Cardiovascular event risk
w10 mg/ml
Late 1990s
(99) (100)
Creatine kinase-myocardial band
CK-MB
Acute myocardial infarct, myocardial necrosis
w1 ng/ml
1960s to 1970s
Cystatin-C
CST3
Cardiovascular event risk
w1 mg/ml
2000s
(101)
Fibrinogen
FBN
Cardiovascular event risk
w1 mg/ml
1980s
(99)
Lipoprotein-associated phospholipase A2
Lp-PLA2
Coronary heart disease risk
w100 ng/ml
2000s
(102) (103)
Myeloperoxidase
MPO
Ischemic heart disease; acute coronary syndrome
w10 ng/ml
1980s
Myoglobin
MYO
Myocardial infarction, necrosis
w10 ng/ml
Late 1970s
(104)
Serum amyloid A
SAA
Coronary artery disease
w10 mg/ml
1990s
(105)
Troponin I
cTnI
Myocardial injury, myocardial infarction
w10 pg/ml
1970s to 1990s
(106)
Troponin T
cTnT
Myocardial injury, myocardial infarction
w10 pg/ml
1970s to 1990s
(107)
*Periods of initial discovery or assay development (108,109).
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
Proteomics in Cardiovascular Research
F I G U R E 1 Sensitivity and Coverage of Various Proteomics Study Designs
Proteomics Approach
Discovery (Shotgun)
Proteome coverage
High - up to 10,000 proteins may be analyzed
Number of samples
Typical sensitivity limit
Low
µg/ml
Medium
ng/ml
Reference concentration in plasma
10-3 (mg/ml)
g/ml Albumin
Example technology Platforms
Notes
High-resolution MS (time-of-flight or Orbitrap instruments)
+ Most unbiased view of proteins
Antibody-based protein arrays
+ Middle ground between scope and sensitivity
LDL 10-6 (µg/ml)
Targeted Discovery
Middle depends on size of target panels or array used
CRP
ng/ml
Aptamer-based protein arrays
Medium pg/ml
CKMB
Data-independent acquisition mass spectrometry
10-9 (ng/ml)
Targeted
Low - typically limited to 10s of proteins
ng/ml
TNF
Multiple-reaction monitoring mass spectrometry (triple-quadruple instruments)
IL-6
ELISA panels
NT-pro BNP
High pg/ml 10-12 (pg/ml)
– Quantification may not be as accurate as targeted approaches
– May require special reagents; target may not be available in commercial panels
+ Excellent specificity and quantification precision – May require method development in specialty laboratories
Properties of 3 common proteomics study designs are listed. Discovery (shotgun), targeted, and targeted discovery proteomics approaches take different strategies to address proteome coverage, number of samples, and sensitivity limits. Discovery proteomics experiments have high coverage (up to 10,000 proteins in a single sample), but have limitations in throughput; fewer samples or subjects can be analyzed. Targeted discovery approaches focus on analyzing a panel of high-potential targets in sufficient numbers of samples. Targeted proteomics is able to achieve the highest sensitivity, which allows the detection of low-abundance plasma markers, such as tumor necrosis factor (TNF) and interleukin (IL)-6, but at the expense of scope and throughput. Graph (center) shows the typical sensitivity range of discovery, targeted discovery, and targeted approaches, juxtaposed with the concentrations of selected plasma proteins and disease markers. Example technological platforms and additional notes are shown on the right. CKMB ¼ creatine kinase-myocardial band; CRP ¼ C-reactive protein; ELISA ¼ enzyme-linked immunosorbent assay; LDL ¼ low-density lipoprotein; MS ¼ mass spectrometry; NT-proBNP ¼ N-terminal pro–B-type natriuretic peptide.
STUDY DESIGNS FOR BIOMARKER
experimental designs. The advantages and disad-
RESEARCH AND DISCOVERY
vantages of technological platforms should therefore be evaluated comprehensively on the basis of the
With precision medicine initiatives demanding a
scope and depth of analysis required, and determined
broader pool of measurable proxies of individual
prior to the commencement of chemical analysis.
characteristics, the call for investigations to discover
Three common designs, all of which can provide both
new candidate biomarkers will likely continue to in-
qualitative and quantitative data on proteins, are: 1)
crease. Successful screens of protein biomarker can-
global discovery; 2) targeted; and 3) targeted discov-
didates require obtaining data: 1) with good coverage
ery. Each has distinct strengths and may be better
of all the proteins present in a sample; and 2) in a
suited to particular experimental goals. Each may be
sufficient number of subjects to ensure adequate
performed in suitable technological platforms that
power for discovery. Unlike microarrays or ribonu-
offer different throughputs, analytical depth, and
cleic acid-sequencing (RNA-seq), a single proteomics
sensitivity. We provide here a brief summary on the
experiment often cannot simultaneously obtain high
strengths
coverage and throughput, necessitating tradeoffs in
approach.
and The
considerations Central
for
Illustration
each
type
presents
of an
2821
2822
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
Proteomics in Cardiovascular Research
C E NT R AL IL L U STR AT IO N Overview of Proteomics Analyses for Cardiovascular Diseases
The Role of Proteomics in the Search For Biomarkers and Mechanisms Of Acquired Cardiovascular Diseases
Sample collection
Data processing and bioinformatics
Data acquisition
Extract proteins from human cohorts or clinical models
Following the acquisitions of large-scale datasets, data are processed:
Three major study approaches:
Cardiac tissue
Data interpretation to extract insights from datasets:
Identify proteins from mass spectra using search engine
Global discovery: Analyze large number of proteins but with lower throughput and sensitivity
Blood/plasma samples
Data interpretation and analysis
Identify interaction networks and/or altered cellular pathways
Deduce relative and/or absolute quantities across samples
Targeted: Analyze fewer proteins but with higher throughput and sensitivity
Connect identified molecular features to phenotypes Recommend identified protein signatures for validation by complementary translational studies
Targeted discovery: Analyze a mid-sized panel of pre-selected proteins Lam, M.P.Y. et al. J Am Coll Cardiol. 2016;68(25):2819–30.
This figure lists major components in common proteomics workflows, as well as their associated experimental considerations in biomarker discovery and disease studies. From top to bottom, protein samples are collected from the plasma of human cohorts or cardiac tissues in animal models according to study goals (either biomarker discovery or mechanistic studies). Three major study approaches (discovery, targeted, and targeted discovery) take different strategies between proteome coverage and analytical throughput, and use different technological platforms, including mass spectrometry and protein arrays. Following the acquisition of largescale datasets, data are processed to identify the protein species present and to deduce their relative quantities across samples. Subsequently, a number of statistical and bioinformatics workflows are used in data interpretation to extract insights from datasets. Network analysis casts proteins in the context of interaction networks or altered cellular pathways. Statistical learning and modeling methods connect the identified molecular features to orthogonal phenotypes, identify signatures, and offer information on subject classification or predictive analysis. The identified protein signatures will require validation, which can be achieved by complementary translational studies, including in vitro biochemical analysis and large cohorts. PTM ¼ post-translational modifications.
overview of data acquisition and analysis; see Table 2
resulting mass spectra are then searched against a
for
protein sequence database (19) or spectral library
a
glossary
of
selected
technological
terms
commonly in use.
(20,21) for protein identification. Various downstream data analyses, such as spectral counting and network
GLOBAL DISCOVERY PROTEOMICS. Global discov-
analysis may be performed to extract other qualita-
ery approaches offer an unbiased view of the protein
tive and quantitative information from the data. A
species that may be found in a sample. Shotgun pro-
major strength of shotgun proteomics is that it casts
teomics is 1 of the most widely used and standardized
the widest net toward potential biomarkers (covering
global discovery strategies. In shotgun analysis, pro-
thousands of proteins) (22) and does not require the
teins are first enzymatically digested (e.g., using
development of individual assays for each target
trypsin) into peptides in vitro, and then subjected to
(23,24). Two weaknesses are its lower overall sensi-
tandem
tivity,
mass
spectrometry
(MS)
analysis.
The
which
may
bias
it
against
biologically
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
2823
Proteomics in Cardiovascular Research
T A B L E 2 Glossary of Selected Technologies Used in Cardiovascular Proteomics
Terms
Description
Amino acid sequence
A segment of the primary structure of the protein where individual amino acids are linked by peptide bonds.
Aptamer array
A non-MS proteomics technology where single-stranded DNA oligonucleotides with affinity for specific protein targets are used as a means to detect and quantify up to w1,000 different protein targets.
Liquid chromatography A common front-end separation technology in proteomics experiments to fractionate complex sample mixture. It is used to resolve and enrich peptides by their chemical properties such as size, hydrophobicity, or charge, and the like. MS
An analytical technology to measure the mass of an analyte (e.g., peptide, protein, amino acid, and so on) in the format of mass-to-charge ratio.
Mass spectrum
The data output of MS. A mass spectrum is a plot displaying the relative intensity or abundance of the analyte ions (y-axis) and their mass-to-charge ratio (x-axis).
Mass/charge (m/z)
The principal measurement of a mass spectrometer: the mass of the ionized analytes divided by the charge they carry. Also commonly referred to as mass-to-charge ratio.
Multiple-reaction monitoring
A targeted proteomics approach in which a particular type of mass spectrometer is programmed to only measure a few peptides and their fragment ions of a target protein of interest, while filtering out all other peptides and proteins in the sample, thus enhancing specificity and sensitivity of the detection/quantification of the targets. Also known as SRM.
Search engine
A software tool used in sequence database search to identify proteins from MS data (e.g., SEQUEST) (110).
Sequence database search
A method to identify proteins from MS data by matching the collected experimental mass spectra to theoretical mass spectra generated from a protein sequence database, such as UniProt (19) or RefSeq (111).
Spectral library search
A method to identify peptides by matching MS data to previously known experimental spectra in a library, such as COPaKB (21) or PeptideAtlas (20).
Shotgun proteomics
A method to identify and quantify proteins on a large scale, in which proteins are first digested into short segments (peptides) that are amenable to sequence determination via tandem MS analysis. The identity of each present protein is then inferred from the sequenced peptides. Also known as bottom-up proteomics.
Tandem mass spectrometry
An analytical method involving the fragmentation of peptides (or other analytes) via multiple stages of MS analyses to obtain sequence/structural information of the analytes. Also known as MS/MS or MS2.
TMT/iTRAQ
Commercially available chemical labels containing stable isotopes, which can be used to label protein samples for multiplexed analysis and comparison.
iTRAQ ¼ isobaric tags for relative and absolute quantitation; MS ¼ mass spectrometry; SRM ¼ selected reaction monitoring; TMT ¼ tandem mass tags.
significant proteins at low abundance (see Improving
for each protein being targeted, which requires sub-
Detection
and
stantial technical expertise from specialized labora-
Throughput section), and less accurate quantification
tories. However, once developed, other investigators
compared with targeted proteomics.
and facilities in clinical and research settings may
TARGETED PROTEOMICS. Targeted proteomics of-
readily adapt the assays. Due to the labor and costs
fers the highest sensitivity and specificity among MS-
required to validate assays, the number of proteins
based proteomics approaches. MS-based targeted
that can be realistically analyzed in a targeted
of
Low-Abundance
Proteins
approaches require the programming of a specific
biomarker search may be limited (w100 or fewer).
instrument (e.g., a triple quadrupole mass spectrom-
Accordingly, targeted proteomics is most suitable for
eter), and development of methods (e.g., multiple
characterizing subsets of likely candidate biomarkers
reaction monitoring) to monitor only a limited num-
with prior disease implications. A limited number of
ber of analytes (via predetermined peptide mass or
non–MS-based antibody assays are also available for
charge and fragment information) to achieve its high
targeted quantification of low-abundance proteins in
sensitivity. It was demonstrated that targeted MS, in
the plasma (e.g., enzyme-linked immunosorbent
conjunction with stable isotope dilution, was able to
assays).
verify a panel of cardiovascular biomarker candidates
TARGETED
at a limit of quantification ranging from 2 to 15 ng/ml
ground between a targeted and an untargeted
(25).
Targeted
MS
is
also
applicable
to
DISCOVERY
APPROACHES. A
middle
post-
approach is sometimes sought, which may be called
translationally modified proteins with PTM site
targeted discovery. The goal is to attain the best of
specificity (26), with the absolute quantity of 14
both worlds: focusing on a finite panel of targets,
phosphorylation sites on cardiac troponin I as a
while maintaining sufficient throughput to analyze a
notable example (27). Ongoing effort is being devoted
large number of samples. Oftentimes, this comes at
to the development of targeted protein assays aimed
the expense of scope (number of proteins analyzed)
at cardiac biomarker analysis (28–30). A disadvantage
and flexibility, but, on the upside, panels of “likely
of targeted proteomics is that the practice of only
bets” can be preselected by manufacturers or by the
using 1 or a few peptides in the experiments may not
investigators to increase the probability of discovery
represent the behavior of the full-length protein (e.g.,
(31). A number of MS-based (e.g., sequential window
due to proteolysis). Specific assays must be developed
acquisition of all theoretical mass spectra) (32) and
2824
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
Proteomics in Cardiovascular Research
non–MS-based affinity proteomics (e.g., antibody-
result quantified >3,000 human plasma proteins in 16
and aptamer-based arrays) (31,33,34), are being used
patients and discovered 333 regulated proteins (38),
to discover candidate biomarkers. Antibody-based
including known markers of spontaneous myocardial
arrays have been used to discover markers for
infarction, such as creatine kinase-myocardial band
ischemic stroke, using a proteomics chip of 92 high-
and troponin. Using 6-plex TMT labels, the proteomic
potential cardiovascular proteins (35), including
analysis of the plasma samples of w1,000 human
B-type natriuretic peptide, growth differentiation
subjects has been recently demonstrated (39). The
factor-15, matrix metalloproteinase-12, and others.
popularization of new 10-plex TMT labels will further
Aptamer arrays that use nucleotides in place of anti-
increase throughput.
bodies for target binding have been used to discover protein myocardial infarct markers and to evaluate a panel of blood proteins as risk factors for prediction of coronary heart diseases (33,34,36,37).
MULTIDIMENSIONAL PROTEIN PARAMETERS IN DISEASE MECHANISM RESEARCH Contemporary proteomics applications in basic dis-
LOW-ABUNDANCE
ease mechanism research often incorporate addi-
PROTEINS AND THROUGHPUT. To improve detec-
tional experimental techniques that target increasing
IMPROVING
DETECTION
OF
for
numbers of “dimensions” of protein properties.
MS-based analysis, commercial immunodepletion
Whereas previous proteomics studies focused largely
columns, including the Human IgY14 and SuperMix
on
columns (Sigma-Aldrich, St. Louis, Missouri), are
regulation in quantity as primary drivers of changes
used to remove top- to medium-abundance proteins.
in function), there has been a growing appreciation of
Simplifying the sample mixture helps unmask low-
other, equally significant proteome parameters that
abundance proteins for detection, but many pro-
modulate cardiovascular biology. These parameters
teins or fragments of interest may be inadvertently
include post-translational regulation, protein-protein
codepleted by the process. Typically, a sample con-
interactions, and protein turnover, each of which may
taining 100 ml of plasma may only yield w140 mg of
be altered in disease and present potential thera-
tion
of
low-abundance
proteins
in
plasma
gene
product
abundance
(up-
and
down-
proteins after 98% of high-abundance species are
peutic targets (9). These parameters are aiding in ef-
depleted, an amount sufficient for a limited number
forts to fully comprehend the disease proteome as a
of replicate analyses. To target challenging proteins
dynamic,
in plasma and other samples, additional preprocess-
velopments in these areas may expand the parameter
ing steps, including size-exclusion chromatography,
space from which future biomarkers may be found.
multidimensional
PROTEIN
liquid
chromatography,
and
multidimensional
entity.
POST-TRANSLATIONAL
Ongoing
de-
REGULATION.
immunocapture may be used, but necessitate greater
Post-translational regulation through covalent modi-
starting amounts of samples (e.g., 0.01 to 1.00 ml of
fication of proteins is a major mechanism of cardiac
plasma) and additional processing time.
signaling. Over 200 protein PTMs that can alter the
To achieve multiplexing, minimize experimental
chemistry and interaction surface of a protein, and
variations, and increase throughput, multiple protein
thereby its structure, activity, binding partners, or
samples can be barcoded with stable isotopes and
subcellular localization, are known (40). Although
analyzed in a single experiment at the expense of
over 500,000 PTM sites are known to exist (41),
costs. Commercially available isobaric tags for rela-
available site-specific antibodies target relatively few
tive and absolute quantitation and tandem mass tag
of them. MS-based proteomics techniques do not
(TMT) labels, small amine-reactive molecules cova-
require separate reagents (i.e., antibodies) for each
lently linked to differential mass reporters and mass
site, and hence can be effectively used to examine
balancers, can be used to tag peptides such that up to
new sites or sites for which antibodies are not
10 sets of barcoded samples may be coanalyzed in 1
available.
MS experiment. During tandem MS, the mass re-
For a number of reasons, PTM studies often require
porters separate from the balancers and act as surro-
additional sample preparation and enrichment con-
gates for comparison of protein quantity of each
siderations (e.g., with titanium dioxide) (42) before
sample. This multiplexing strategy has been profit-
data acquisition. First, modified proteins frequently
ably used to identify potential infarct markers in a
exist in lower abundance than the total protein pop-
study that analyzed the plasma of 16 patients at
ulation (although in some instances, such as in the
multiple time points following septal ablation treat-
case of secreted glycoproteins, almost every protein
ment. With the aid of 4-plex isobaric tags for relative
molecule is modified). For example, if only 10% of a
and absolute quantitation labels, the proteomics
protein species is modified, then the modified
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
Proteomics in Cardiovascular Research
proteins are effectively 10 times more difficult to
histones. A family of enzymes known as histone
detect than the total (modified plus unmodified)
acetyltransferases catalyzes the addition of the acetyl
population. Second, whereas identifying a protein
group to proteins, whereas histone deacetylases
only requires detection of any of its multiple pro-
revert the modification. Although protein acetylation
teolytic peptides along its primary structure, PTM
was classically described on histones, other proteins
analysis requires the precise detection of the peptide
are also modified. Protein acetylation participates in
containing the PTM site, which sometimes can lie in a
nonchromatin processes, including metabolism and
region of the protein that is not amenable to MS
protein localization, as well as protein stability
detection (e.g., sequences without trypsin digestion
(45,46), and can be mapped with proteomics tech-
sites). Third, many PTMs are chemically labile and
niques following enrichment by anti–acetyl-lysine
biologically transient, making their capture and
antibodies (62,63). Over one-third of mitochondrial
enrichment before signal measurement essential.
proteins are subjected to acetylation, including a
PTMs under intense investigation for their roles in cardiovascular (43,44),
diseases
acetylation
include
(45,46),
phosphorylation
SUMOylation
large number of metabolic enzymes. Acetylation in mitochondria may function in cardioprotective con-
(47),
texts (46), whereas hyperacetylation of succinate
glycosylation (48,49), S-nitrosylation (50,51), ubiq-
dehydrogenase in mitochondria may drive energy
uitination (52), and others (53,54). The remainder of
imbalance in the development of heart failure (63).
this section highlights proteomics approaches to un-
Reactive
oxygen
and
nitrogen
species
can
derstand protein phosphorylation, acetylation, and
confer covalent modifications (e.g., S-nitrosylation,
oxidative modifications.
S-sulfhydration, and S-glutathionylation) on cys-
Multiple major kinase pathways operate in the
teine residues in redox-sensitive proteins. These
heart, including protein kinase A, protein kinase B,
modifications can change the activity and binding
mitogen-activated protein kinase or extracellular
surfaces of target proteins, and thus directly partic-
signal-regulated kinase, protein kinase C, glycogen
ipate in cardiovascular biology in a number of dis-
synthase kinase 3b, calcium/calmodulin-dependent
ease contexts. For example, it is known that nitric
protein kinase II, and others; hence, it is no sur-
oxide serves as a critical signaling messenger in
prise that phosphoproteomics is commonly applied
ischemic preconditioning. Nitric oxide signals partly
in the study of heart diseases. A well-known
by causatively increasing protein S-nitrosylation,
example of a phosphorylated protein essential for
which
cardiac energetics is pyruvate dehydrogenase (PDH)
dioprotection by reducing other, pernicious forms of
(55), which converts pyruvate to acetyl coenzyme A
cysteine oxidation on the target proteins during
for respiration, and whose activity is dictated by its
reperfusion
phosphorylation by PDH kinases. PDH phosphoryla-
modification en masse, biotin switch proteomics are
tion thus serves as a major regulatory checkpoint of
used that first chemically block free thiols, then uses
cardiac energy metabolism. Classically, phosphory-
a specific reducing agent to reduce different reduc-
lation sites were mapped by phosphorus 32 labeling
tive states of oxidative PTMs (e.g., ascorbic acids
and Edman degradation. Up to w50,000 phosphor-
may be used to reduce only S-nitrosylated thiols, but
has
been
injury
proposed
(64,65).
to
To
mediate
detect
car-
oxidative
ylation sites may now be identified in a single study
not other modifications), and finally labels the freed
using
thiols with biotin or isotope-labeled tags (cys-TMT or
shotgun
proteomics
(56)
combined
with
enrichment strategies. Mapping in the heart has
iodo-TMT) (51,66).
unearthed a large number of phosphorylation sites, including those common to multiple tissues and
PROTEIN TEMPORAL DYNAMICS. Protein homeosta-
many that are unique to the cardiovascular system
sis and proteolysis are broadly implicated in heart
(57). Targeted mapping efforts may also be directed
diseases and injuries. Classical studies of protein
toward selected cellular compartments (e.g., mito-
turnover required pulse-chase of radioisotopes and
chondria) (58), pathways (e.g., those targeted by
isolation of target proteins. With recent advances,
phosphodiesterase 9A/phosphodiesterase 5, calcium/
proteomics studies can measure turnover dynamics
calmodulin-dependent protein kinase II) (59,60),
of thousands of proteins in multiple tissues and or-
specific proteins (e.g., the myosin-binding protein C
ganisms (67,68). Combining stable isotope labeling,
phosphoproteome) (61), or multiple specific sites on
mathematical modeling, and shotgun proteomics,
the same proteins (26). Acetylation of histones plays a key role in gene
recent investigations into the turnover of w5,000 cardiac
proteins
in
mouse
cardiac
hypertrophy
expression by modulating the accessibility of tran-
(69,70) revealed novel correlations between prote-
scriptional elements to DNA wound around the
ome profiles and phenotypic functional limitations.
2825
2826
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
Proteomics in Cardiovascular Research
For example, although the hypertrophic heart is
INTERPRETATION AND VALIDATION OF
known to switch in fuel preference from fatty acid to
PROTEOMICS DATA
glucose, the expression of glycolytic proteins does not change accordingly (71). Instead, the turnover of
Several considerations are provided here regarding
most glycolytic proteins was found to accelerate
how to interpret proteomic results, as well as how to
drastically
use new workflows and technologies.
in
hypertrophy
without
concomitant
expression changes. Thus static expression profiling
SCOPE AND SENSITIVITY. With incomplete coverage,
experiments alone would not have discovered this
the old adage that the absence of evidence is not
aspect of cardiac pathophysiology had there been no a
evidence of absence is especially true for proteomics
priori knowledge of increased glycolysis, whereas a
experiments. The number of proteins and peptides in
large-scale experiment of protein turnover might
a shotgun experiment is an important indicator of
have led to the generation of relevant hypotheses. A
the depth of coverage in data acquisition. Hence, if
possible connection between turnover and function is
a known biomarker is not reported or reproduced in
that old proteins may have reduced activity compared
a discovery-based experiment in the cohort, a perti-
with young, functional proteins. For example, the
nent question is whether the technological platform
glycolysis enzyme triose-phosphate isomerase can
used has sufficient sensitivity and precision to sup-
become
port reproducible measurements at endogenous
chemically
deamidated
upon
catalytic
cycling. In another example, the stem cell factor or KIT ligand has been found to lose biological potency
concentration. The predictive value of a putative biomarker de-
by 50-fold after chemical deamidation (72).
pends not only on the magnitude of its difference in
PROTEIN-PROTEIN INTERACTIONS. Physical liaison
normal versus diseased or at-risk individuals in the
between proteins is a primary means whereby infor-
population, but also on the effect size, defined as
mation is transduced along signaling pathways. The
the number of SDs between 2 measured means.
target specificity and activity of protein kinase A are
Hence, for the purpose of protein biomarker dis-
markedly regulated by which of the w40 A-kinase
covery, the coefficient of variation of measurement
anchoring proteins it primarily interacts with (73), to
is essential for determining whether there is suffi-
mediate kinase signaling, and which can change
cient power to distinguish significant differences
during heart disease (74). Protein-protein interaction
between healthy and diseased samples. Beyond the
analysis is pursued to characterize signaling net-
precision characteristics of the technological plat-
works, discover the components of protein com-
forms, sample processing, as well as biological var-
plexes, and identify the contexts in which a protein
iations in human cohorts (both intrasubject and
may function in health and disease (75). A powerful
intersubject), may also impart significant variance in
approach to analyze protein-protein interaction is
protein measurements. Thus, individual putative
affinity purification-MS (AP-MS) (76,77), which pre-
protein markers often require lengthy verification
cipitates bait proteins, along with their interacting
and validation pipelines that involve large cohorts
partners, using antibodies or another purification
(83). Proteomics assays have reported typical co-
method. Using unbiased AP-MS, a recent study
efficients of variation in the range of <5% for
discovered the association of the cardiac transcrip-
intralaboratory measurements (34,84) to <20% for
tion factor TBX5 with the nucleosome remodeling
interlaboratory measurements (85). Other measure-
deacetylase repressor complex, revealing a novel
ments of analytical performance that can have a
mechanism for TBX5 in cardiac development and
drastic impact on the likelihood of payoff in down-
congenital heart diseases (78). Although it is the
stream verification and validation processes include
current mainstay of interactome research, AP-MS
the limit of detection, limit of quantification, and
specificity can vary. Even with careful washing,
linearity of measurement, which are covered at
nonspecific binding occurs and contaminants can
length elsewhere (83).
remain. Thus, identified interactors should be veri-
FALSE DISCOVERY RATE. As in other large-scale ap-
fied by complementary methods. New methods are
proaches, both the number of identified proteins and
currently being developed to ameliorate this chal-
the significance of quantitative comparisons in a
lenge, including in silico filtering of interactome data
quantitative proteomics experiment may become in-
(79,80) and cross-linking MS (81). The latter uses
flated if controls for false discovery rates (FDRs) are
reactive chemicals to physically link together pro-
not adequate. In protein identification, the FDR of a
teins in apposition for confident MS identification
database search provides an estimate for the propor-
(reviewed in Leitner et al. [82]).
tion of incorrect protein identification in the result list.
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
Proteomics in Cardiovascular Research
An FDR of 1% is commonly deemed acceptable,
procedures, or in peripheral blood versus proximal
meaning that in a profiling experiment claiming to
tissue fluids, such as coronary sinus effluents, to filter
identify 5,000 proteins, up to 50 may be mis-
out spurious associations. An in-depth discourse on
identifications. FDR is sometimes calculated with the
biomarker verification and validation can be found
aid of decoy databases, which contain scrambled or
elsewhere (83).
reversed protein sequences that are compared to experimental data, along with real protein sequences. The number of identified decoy sequences provides an estimate of the number of false discoveries among regular database sequence matches. For quantitative proteomics comparisons between samples, as many hypotheses as the number of proteins compared are being tested per sample pair. Multiple-testing corrections (e.g., Bonferroni corrections) are used to control for type I errors (incorrectly rejecting the null hypothesis) that arise from making multiple comparisons (1 per protein per sample pair). However, it must be noted that even infinitesimal p values are no indication that results are biomedically important (86), and do not obviate the need to validate with follow-up studies, including in vitro experimentation or in cohorts.
OUTLOOK With continued technological progress, new interactions of proteomics with genetics and genomics are now becoming possible. Proteomics studies applied to large cohorts can be integrated with genetic polymorphisms to identify protein quantitative trait loci (pQTL) that control protein abundance, and perhaps thereby control phenotypic traits. Proteogenomics studies, combining RNA-seq and proteomics data from common datasets, have been undertaken in cancer research to identify the protein coding consequences of cancer variants and mutations (91), whereas a recent multiomics study in 80 mouse genetic backgrounds demonstrated that multiple types of omics data can complement each other to characterize
COMPARISON
WITH
EXISTING
mitochondrial
functional
regulation
(92).
TECHNIQUES. In
Further integration of proteomics and genomics
addition to technical considerations, criteria to
methods will allow examination of nonstandard gene
nominate a differentially expressed protein as a
products, including short open reading frames, un-
candidate biomarker can vary. Currently, no gold
annotated transcripts, and alternative splicing iso-
standard exists for what constitutes a good candidate
form proteins. By greatly expanding the depth of the
(87). Decision justifications in published reports range
proteome that may be experimentally accessed, the
from formal statistical inferences of individual pro-
development of isoform proteomics, in particular,
teins (88), panels of proteins after feature selection
will have important implications for cardiovascular
(89), to additional criteria, such as cutoffs in the raw
research. These isoforms may present with differen-
magnitude of fold change (e.g., >5-fold changes) in at
tial expression levels or patterns, localizations, in-
least n number of patients (90), to more custom
teractions, and PTMs in different cell types and
modeling that adjust for covariates in risk assess-
during disease progression, including in ischemic
ments (31,35). In determining whether the presented
cardiomyopathy (93) and hypertrophy (94). Although
candidates are worthy of prioritized verification
RNA-seq has discovered many alternative isoforms at
studies, it stands to reason that the results of dis-
the transcript level, the majority of isoform tran-
covery experiments should be carefully scrutinized
scripts have not been validated at the protein level.
for both nominal significance (e.g., p values), and for
Many alternatively spliced transcripts may lead to
the magnitude and variance of biological changes
frame-shifts and be targeted by nonsense-mediated
(effect size and confidence interval).
decay. The functional significance of many tran-
Ultimately, a candidate biomarker or protein with
script isoforms may soon be addressed by proteomics
potential biomedical significance needs to be validated
using custom sequence databases translated from
by orthogonal approaches in independent cohorts or
RNA-seq transcripts.
models. In silico methods may be used to triage a list of
Broader applications will also be facilitated by
differentially expressed proteins and prioritize candi-
continued
dates for validation, for example, by determining their
coverage. For example, newly developed neutron
improvements
in
throughput
and
tissue expression specificity and half-lives. Promising
encoding (NeuCode) techniques will allow over 30
biomarkers may be further credentialed with targeted
samples to be combined for MS analysis (95), leading
discovery approaches that quantify the status of the
to a 5-fold increase in throughput for investigating
candidates in larger numbers of subjects and condi-
large cohorts. With new affinity proteomics plat-
tions. For instance, protein profiles may be compared
forms, such as protein and aptamer arrays, we have
in
already seen further development of preselected
paired
controls
before
and
after
unrelated
2827
2828
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
Proteomics in Cardiovascular Research
cardiovascular-specific protein panels (33), which
innovative breakthroughs in cardiovascular medicine
could allow the disease status of the likeliest candi-
for years to come.
dates to be screened across large sample pools. In summary, with core proteomics technologies having
REPRINT REQUESTS AND CORRESPONDENCE: Dr.
now matured to provide high protein and sample
Maggie P.Y. Lam, David Geffen School of Medicine at
coverage, new applications are increasingly available
UCLA, Department of Physiology & Bioinformatics,
to interrogate the dynamic parameters that connect
675 Charles E. Young Drive, MRL Building, Suite 1-
gene expression to physiological functions. We envi-
625, Los Angeles, California 90095. E-mail: magelpy@
sion that proteomics-based discovery-driven plat-
ucla.edu. OR Dr. Elizabeth Murphy, National Heart,
forms
molecular
Lung, and Blood Institute, NIH, Building 10, Room
phenotyping of individuals in the era of precision
8N202, 10 Center Drive, Bethesda, Maryland 20892.
medicine, and will provide ample opportunities for
E-mail:
[email protected].
will
continue
to
support
REFERENCES 1. McDonough JL, Labugger R, Pickett W, et al. Cardiac troponin I is modified in the myocardium of bypass patients. Circulation 2001;103:58–64. 2. Labugger R, Organ L, Collier C, et al. Extensive troponin I and T modification detected in serum from patients with acute myocardial infarction. Circulation 2000;102:1221–6. 3. Weekes J, Wheeler CH, Yan JX, et al. Bovine dilated cardiomyopathy: proteomic analysis of an animal model of human dilated cardiomyopathy. Electrophoresis 1999;20:898–906. 4. Ping P, Zhang J, Pierce WM, et al. Functional proteomic analysis of protein kinase C ε signaling complexes in the normal heart and during cardioprotection. Circ Res 2001;88:59–62. 5. Edmondson RD, Vondriska TM, Biederman KJ, et al. Protein kinase C ε signaling complexes include metabolismand transcription/ translation-related proteins: complimentary separation techniques with LC/MS/MS. Mol Cell Proteomics 2002;1:421–33. 6. Lindsey ML, Goshorn DK, Comte-Walters S, et al. A multidimensional proteomic approach to identify hypertrophy-associated proteins. Proteomics 2006;6:2225–35. 7. Sun J, Picht E, Ginsburg KS, et al. Hypercontractile female hearts exhibit increased S-nitrosylation of the L-type Ca2þ channel a1 subunit and reduced ischemia/reperfusion injury. Circ Res 2006;98:403–11. 8. Ge Y, Rybakova IN, Xu Q, et al. Top-down highresolution mass spectrometry of cardiac myosin binding protein C revealed that truncation alters protein phosphorylation state. Proc Natl Acad Sci U S A 2009;106:12658–63. 9. Larance M, Lamond AI. Multidimensional proteomics for cell biology. Nat Rev Mol Cell Biol 2015;16:269–80. 10. Liu Y, Beyer A, Aebersold R. On the dependency of cellular protein levels on mRNA abundance. Cell 2016;165:535–50. 11. Vogel C, Abreu Rde S, Ko D, et al. Sequence signatures and mRNA concentration can explain two-thirds of protein abundance variation in a human cell line. Mol Syst Biol 2010;6:400. 12. Tian Q, Stepaniants SB, Mao M, et al. Integrated genomic and proteomic analyses of gene
expression in mammalian cells. Mol Cell Proteomics 2004;3:960–9. 13. Schwanhäusser B, Busse D, Li N, et al. Global quantification of mammalian gene expression control. Nature 2011;473:337–42. 14. Lundberg E, Fagerberg L, Klevebring D, et al. Defining the transcriptome and proteome in three functionally different human cell lines. Mol Syst Biol 2010;6:450. 15. Marshall KD, Edwards MA, Krenz M, et al. Proteomic mapping of proteins released during necrosis and apoptosis from cultured neonatal cardiac myocytes. Am J Physiol Cell Physiol 2014; 306:C639–47. 16. Van Eyk JE. Proteomics: unraveling the complexity of heart disease and striving to change cardiology. Curr Opin Mol Ther 2001;3:546–53.
patient plasma by targeted mass spectrometry and stable isotope dilution. Mol Cell Proteomics 2009;8:2339–49. 26. Lam MP, Lau E, Scruggs SB, et al. Site-specific quantitative analysis of cardiac mitochondrial protein phosphorylation. J Proteomics 2013;81: 15–23. 27. Zhang P, Kirk JA, Ji W, et al. Multiple reaction monitoring to identify site-specific troponin I phosphorylated residues in the failing human heart. Circulation 2012;126:1828–37. 28. Percy AJ, Yang J, Hardie DB, et al. Precise quantitation of 136 urinary proteins by LC/MRMMS using stable isotope labeled peptides as internal standards for biomarker discovery and/or verification studies. Methods 2015;81:24–33.
Plasma Proteome Database as a resource for proteomics research: 2014 update. Nucleic Acids Res 2014;42:D959–65.
29. Fu Q, Chen Z, Zhang S, et al. Multiple and selective reaction monitoring using triple quadrupole mass spectrometer: preclinical large cohort analysis. Methods Mol Biol 2016;1410: 249–64.
18. Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol Cell Proteomics 2002;1: 845–67.
30. Percy AJ, Byrns S, Chambers AG, Borchers CH. Targeted quantitation of CVD-linked plasma proteins for biomarker verification and validation. Expert Rev Proteomics 2013;10:567–78.
19. Pundir S, Magrane M, Martin MJ, et al. Searching and navigating UniProt databases. Curr Protoc Bioinformatics 2015;50:1.27.1–1.27.10.
31. Gerstein HC, Paré G, McQueen MJ, et al.,
17. Nanjappa V, Thomas JK, Marimuthu A, et al.
20. Kusebauch U, Deutsch EW, Campbell DS, et al. Using PeptideAtlas, SRMAtlas, and PASSEL: comprehensive resources for discovery and targeted proteomics. Curr Protoc Bioinformatics 2014;46:13.25.1–28. 21. Zong N, Li H, Li H, et al. Integration of cardiac proteome biology and medicine by a specialized knowledgebase. Circ Res 2013;113:1043–53. 22. Riley NM, Hebert AS, Coon JJ. Proteomics moves into the fast lane. Cell Syst 2016;2:142–3. 23. Bensimon A, Heck AJ, Aebersold R. Mass spectrometry-based proteomics and network biology. Annu Rev Biochem 2012;81:379–405. 24. Zhang Y, Fonslow BR, Shan B, et al. Protein analysis by shotgun/bottom-up proteomics. Chem Rev 2013;113:2343–94. 25. Keshishian H, Addona T, Burgess M, et al. Quantification of cardiovascular biomarkers in
Outcome Reduction With Initial Glargine Intervention Trial Investigators. Identifying novel biomarkers for cardiovascular events or death in people with dysglycemia. Circulation 2015;132: 2297–304. 32. Sajic T, Liu Y, Aebersold R. Using dataindependent, high-resolution mass spectrometry in protein biomarker research: perspectives and clinical applications. Proteomics Clin Appl 2015;9: 307–21. 33. Ngo D, Sinha S, Shen D, et al. Aptamer-based proteomic profiling reveals novel candidate biomarkers and pathways in cardiovascular disease. Circulation 2016;134:270–85. 34. Ganz P, Heidecker B, Hveem K, et al. Development and validation of a protein-based risk score for cardiovascular outcomes among patients with stable coronary heart disease. JAMA 2016; 315:2532–41. 35. Lind L, Siegbahn A, Lindahl B, et al. Discovery of new risk markers for ischemic stroke using a
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
Proteomics in Cardiovascular Research
novel targeted proteomics chip. Stroke 2015;46: 3340–7.
with cysteine-reactive tandem mass tags: short communication. Circ Res 2012;111:1308–12.
CysTMT6 switch assay. Methods Mol Biol 2013; 1005:169–79.
36. Sabatine MS. Using aptamer-based technology to probe the plasma proteome for cardiovascular disease prediction. JAMA 2016;315:2525–6.
52. Zong N, Ping P, Lau E, et al. Lysine ubiquitination and acetylation of human cardiac 20S proteasomes. Proteomics Clin Appl 2014;8:590–4.
37. Gramolini A, Lau E, Liu PP. Identifying lowabundance biomarkers: aptamer-based prote-
53. Fert-Bober J, Giles JT, Holewinski RJ, et al. Citrullination of myofilament proteins in heart
67. Kim TY, Wang D, Kim AK, et al. Metabolic labeling reveals proteome dynamics of mouse mitochondria. Mol Cell Proteomics 2012;11: 1586–94.
omics potentially enables more sensitive detection in cardiovascular diseases. Circulation 2016;134: 286–9.
failure. Cardiovasc Res 2015;108:232–42.
38. Keshishian H, Burgess MW, Gillette MA, et al. Multiplexed, quantitative workflow for sensitive biomarker discovery in plasma yields novel candidates for early myocardial injury. Mol Cell Proteomics 2015;14:2375–93. 39. Cominetti O, Nuñez Galindo A, Corthésy J, et al. Proteomic biomarker discovery in 1000 human plasma samples with mass spectrometry. J Proteome Res 2016;15:389–99. 40. Olsen JV, Mann M. Status of large-scale analysis of post-translational modifications by mass spectrometry. Mol Cell Proteomics 2013;12: 3444–52. 41. Huang KY, Su MG, Kao HJ, et al. dbPTM 2016: 10-year anniversary of a resource for posttranslational modification of proteins. Nucleic Acids Res 2016;44:D435–46. 42. Thingholm TE, Jørgensen TJ, Jensen ON, et al. Highly selective enrichment of phosphorylated peptides using titanium dioxide. Nat Protoc 2006; 1:1929–35. 43. Lundby A, Andersen MN, Steffensen AB, et al. In vivo phosphoproteomics analysis reveals the cardiac targets of b-adrenergic receptor signaling. Sci Signal 2013;6:rs11. 44. Schechter MA, Hsieh MK, Njoroge LW, et al. Phosphoproteomic profiling of human myocardial tissues distinguishes ischemic from non-ischemic end stage heart failure. PLoS One 2014;9:e104157. 45. Karamanlidis G, Lee CF, Garcia-Menendez L, et al. Mitochondrial complex I deficiency increases protein acetylation and accelerates heart failure. Cell Metab 2013;18:239–50. 46. Nguyen TT, Wong R, Menazza S, et al. Cyclophilin D modulates mitochondrial acetylome. Circ Res 2013;113:1308–19. 47. Yang W, Paschen W. SUMO proteomics to decipher the SUMO-modified proteome regulated by various diseases. Proteomics 2015;15:1181–91. 48. Barallobre-Barreiro J, Gupta SK, Zoccarato A, et al. Glycoproteomics reveals decorin peptides with anti-myostatin activity in human atrial fibrillation. Circulation 2016;134:817–32. 49. Parker BL, Palmisano G, Edwards AV, et al. Quantitative N-linked glycoproteomics of myocardial ischemia and reperfusion injury reveals
54. Lindsey ML, Iyer RP, Zamilpa R, et al. A novel collagen matricryptin reduces left ventricular dilation post-myocardial infarction by promoting scar formation and angiogenesis. J Am Coll Cardiol 2015;66:1364–74. 55. Kerbey AL, Randle PJ, Cooper RH, et al. Regulation of pyruvate dehydrogenase in rat heart. Mechanism of regulation of proportions of dephosphorylated and phosphorylated enzyme by oxidation of fatty acids and ketone bodies and of effects of diabetes: role of coenzyme A, acetylcoenzyme A and reduced and oxidized nicotinamide-adenine dinucleotide. Biochem J 1976;154:327–48. 56. Sharma K, D’Souza RC, Tyanova S, et al. Ultradeep human phosphoproteome reveals a distinct regulatory nature of Tyr and Ser/Thrbased signaling. Cell Rep 2014;8:1583–94. 57. Lundby A, Secher A, Lage K, et al. Quantitative maps of protein phosphorylation sites across 14 different rat organs and tissues. Nat Commun 2012;3:876. 58. Deng N, Zhang J, Zong C, et al. Phosphoproteome analysis reveals regulatory sites in major pathways of cardiac mitochondria. Mol Cell Proteomics 2011;10. M110.000117. 59. Lee DI, Zhu G, Sasaki T, et al. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease. Nature 2015;519: 472–6. 60. Scholten A, Preisinger C, Corradini E, et al. Phosphoproteomics study based on in vivo inhibition reveals sites of calmodulin-dependent protein kinase II regulation in the heart. J Am Heart Assoc 2013;2:e000318. 61. Kooij V, Holewinski RJ, Murphy AM, et al. Characterization of the cardiac myosin binding protein-C phosphoproteome in healthy and failing human hearts. J Mol Cell Cardiol 2013;60:116–20. 62. Svinkina T, Gu H, Silva JC, et al. Deep, quantitative coverage of the lysine acetylome using novel anti-acetyl-lysine antibodies and an optimized proteomic workflow. Mol Cell Proteomics 2015;14:2429–40. 63. Horton JL, Martin OJ, Lai L, et al. Mitochondrial protein hyperacetylation in the failing heart. JCI Insight 2016;2:e84897. 64. Sun J, Aponte AM, Menazza S, et al. Additive cardioprotection by pharmacological post-
early remodeling in the extracellular environment. Mol Cell Proteomics 2011;10. M110.006833.
conditioning with hydrogen sulfide and nitric oxide donors in mouse heart: S-sulfhydration vs. Snitrosylation. Cardiovasc Res 2016;110:96–106.
50. Wang SB, Foster DB, Rucker J, et al. Redox regulation of mitochondrial ATP synthase: implications for cardiac resynchronization therapy. Circ Res 2011;109:750–7.
65. Sun J, Nguyen T, Aponte AM, et al. Ischaemic preconditioning preferentially increases protein Snitrosylation in subsarcolemmal mitochondria. Cardiovasc Res 2015;106:227–36.
51. Kohr MJ, Aponte A, Sun J, et al. Measurement of S-nitrosylation occupancy in the myocardium
66. Murray CI, Chung HS, Uhrigshardt H, et al. Quantification of mitochondrial S-nitrosylation by
68. Price JC, Holmes WE, Li KW, et al. Measurement of human plasma proteome dynamics with 2H2O and liquid chromatography tandem mass spectrometry. Anal Biochem 2012;420:73–83. 69. Lam MP, Wang D, Lau E, et al. Protein kinetic signatures of the remodeling heart following isoproterenol stimulation. J Clin Invest 2014;124: 1734–44. 70. Lau E, Cao Q, Ng DC, et al. A large dataset of protein dynamics in the mammalian heart proteome. Sci Data 2016;3:160015. 71. Kolwicz SC Jr., Tian R. Glucose metabolism and cardiac hypertrophy. Cardiovasc Res 2011;90: 194–201. 72. Hsu YR, Chang WC, Mendiaz EA, et al. Selective deamidation of recombinant human stem cell factor during in vitro aging: isolation and characterization of the aspartyl and isoaspartyl homodimers and heterodimers. Biochemistry 1998;37: 2251–62. 73. Burgers PP, van der Heyden MA, Kok B, et al. A systematic evaluation of protein kinase A-A-kinase anchoring protein interaction motifs. Biochemistry 2015;54:11–21. 74. Aye TT, Soni S, van Veen TA, et al. Reorganized PKA-AKAP associations in the failing human heart. J Mol Cell Cardiol 2012;52:511–8. 75. Pankow S, Bamberger C, Calzolari D, et al.
DF508 CFTR interactome remodelling promotes rescue of cystic fibrosis. Nature 2015;528:510–6. 76. Morris JH, Knudsen GM, Verschueren E, et al. Affinity purification-mass spectrometry and network analysis to understand protein-protein interactions. Nat Protoc 2014;9:2539–54. 77. Huttlin EL, Ting L, Bruckner RJ, et al. The BioPlex network: a systematic exploration of the human interactome. Cell 2015;162:425–40. 78. Waldron L, Steimle JD, Greco TM, et al. The cardiac TBX5 interactome reveals a chromatin remodeling network essential for cardiac septation. Dev Cell 2016;36:262–75. 79. Rizzetto S, Priami C, Csikász-Nagy A. Qualitative and quantitative protein complex prediction through proteome-wide simulations. PLoS Comput Biol 2015;11:e1004424. 80. Goldfarb D, Hast BE, Wang W, et al. Spotlite: web application and augmented algorithms for predicting co-complexed proteins from affinity purification–mass spectrometry data. J Proteome Res 2014;13:5944–55. 81. Gingras AC, Gstaiger M, Raught B, et al. Analysis of protein complexes using mass spectrometry. Nat Rev Mol Cell Biol 2007;8:645–54. 82. Leitner A, Faini M, Stengel F, et al. Crosslinking and mass spectrometry: an integrated technology to understand the structure and function of molecular machines. Trends Biochem Sci 2016;41:20–32.
2829
2830
Lam et al.
JACC VOL. 68, NO. 25, 2016 DECEMBER 27, 2016:2819–30
Proteomics in Cardiovascular Research
83. Rifai N, Gillette MA, Carr SA. Protein biomarker discovery and validation: the long and uncertain path to clinical utility. Nat Biotechnol 2006;24: 971–83. 84. Percy AJ, Chambers AG, Yang J, et al. Multiplexed MRM-based quantitation of candidate cancer biomarker proteins in undepleted and nonenriched human plasma. Proteomics 2013;13: 2202–15. 85. Abbatiello SE, Schilling B, Mani DR, et al. Large-scale interlaboratory study to develop, analytically validate and apply highly multiplexed, quantitative peptide assays to measure cancerrelevant proteins in plasma. Mol Cell Proteomics 2015;14:2357–74. 86. Goodman SN. Aligning statistical and scientific reasoning. Science 2016;352:1180–1. 87. de Lemos JA, Rohatgi A, Ayers CR. Applying a big data approach to biomarker discovery: running before we walk? Circulation 2015;132:2289–92. 88. Keshishian H, Burgess MW, Gillette MA, et al. Multiplexed, quantitative workflow for sensitive biomarker discovery in plasma yields novel candidates for early myocardial injury. Mol Cell Proteomics 2015;14:1–45.
93. Kong SW, Hu YW, Ho JW, et al. Heart failureassociated changes in RNA splicing of sarcomere genes. Circ Cardiovasc Genet 2010;3:138–46. 94. Song HK, Hong SE, Kim T, et al. Deep RNA sequencing reveals novel cardiac transcriptomic signatures for physiological and pathological hypertrophy. PLoS One 2012;7:e35552. 95. Hebert AS, Merrill AE, Bailey DJ, et al. Neutron-encoded mass signatures for multiplexed proteome quantification. Nat Methods 2013;10: 332–4. 96. Stubbs P, Seed M, Lane D, et al. Lipoprotein(a) as a risk predictor for cardiac mortality in patients with acute coronary syndromes. Eur Heart J 1998; 19:1355–64. 97. McQueen MJ, Hawken S, Wang X, et al., INTERHEART study investigators. Lipids, lipoproteins, and apolipoproteins as risk markers of myocardial infarction in 52 countries (the INTERHEART study): a case-control study. Lancet 2008; 372:224–33. 98. de Lemos JA, McGuire DK, Drazner MH. Btype natriuretic peptide in cardiovascular disease. Lancet 2003;362:316–22.
89. Yin X, Subramanian S, Hwang SJ, et al. Protein biomarkers of new-onset cardiovascular disease:
99. Emerging Risk Factors Collaboration. C-reactive protein, fibrinogen, and cardiovascular disease prediction. N Engl J Med 2012;367:1310–20.
prospective study from the systems approach to biomarker research in cardiovascular disease initiative. Arterioscler Thromb Vasc Biol 2014;34: 939–45.
100. Puleo PR, Guadagno PA, Roberts R, et al. Early diagnosis of acute myocardial infarction based on assay for subforms of creatine kinaseMB. Circulation 1990;82:759–64.
90. Addona TA, Shi X, Keshishian H, et al. A pipeline that integrates the discovery and verification of plasma protein biomarkers reveals candidate markers for cardiovascular disease. Nat Biotechnol 2011;29:635–43.
101. Shlipak MG, Sarnak MJ, Katz R, et al. Cystatin C and the risk of death and cardiovascular events among elderly persons. N Engl J Med 2005;352: 2049–60.
91. Mertins P, Mani DR, Ruggles KV, et al. Proteogenomics connects somatic mutations to signalling in breast cancer. Nature 2016;534: 55–62. 92. Williams EG, Wu Y, Jha P, et al. Systems proteomics of liver mitochondria function. Science 2016;352:aad0189.
102. Daniels LB, Laughlin GA, Sarno MJ, et al. Lipoprotein-associated phospholipase A2 is an independent predictor of incident coronary heart disease in an apparently healthy older population: the Rancho Bernardo Study. J Am Coll Cardiol 2008;51:913–9. 103. Brennan ML, Penn MS, Van Lente F, et al. Prognostic value of myeloperoxidase in patients
with chest pain. N Engl J Med 2003;349: 1595–604. 104. Kavsak PA, MacRae AR, Newman AM, et al. Effects of contemporary troponin assay sensitivity on the utility of the early markers myoglobin and CKMB isoforms in evaluating patients with possible acute myocardial infarction. Clin Chim Acta 2007;380:213–6. 105. Johnson BD, Kip KE, Marroquin OC, et al. Serum amyloid A as a predictor of coronary artery disease and cardiovascular outcome in women: the National Heart, Lung, and Blood InstituteSponsored Women’s Ischemia Syndrome Evaluation (WISE). Circulation 2004;109:726–32. 106. Adams JE III, Bodor GS, Dávila-Román VG, et al. Cardiac troponin I. A marker with high specificity for cardiac injury. Circulation 1993;88: 101–6. 107. Reichlin T, Hochholzer W, Bassetti S, et al. Early diagnosis of myocardial infarction with sensitive cardiac troponin assays. N Engl J Med 2009; 361:858–67. 108. Danese E, Montagnana M. An historical approach to the diagnostic biomarkers of acute coronary syndrome. Ann Transl Med 2016;4:194. 109. Ladenson JH. A personal history of markers of myocyte injury [myocardial infarction]. Clin Chim Acta 2007;381:3–8. 110. Eng JK, McCormack AL, Yates JR. An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J Am Soc Mass Spectrom 1994;5: 976–89. 111. O’Leary NA, Wright MW, Brister JR, et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res 2016;44: D733–45.
KEY WORDS mass spectrometry, molecular phenotyping, post-translational modifications, protein arrays, protein dynamics, protein signatures