You are on page 1of 12

Review

pubs.acs.org/ac

High Resolution Mass Spectrometry


Feng Xian,† Christopher L. Hendrickson,*,†,‡ and Alan G. Marshall*,†,‡

Department of Chemistry and Biochemistry, Florida State University, 95 Chieftain Way, Tallahassee, Florida 32310-4390,
United States

Ion Cyclotron Resonance Program, National High Magnetic Field Laboratory, 1800 East Paul Dirac Drive, Tallahassee,


Florida 32310-4005, United States
CONTENTS resolving power is required to distinguish the same mass difference
Downloaded via BENEMERITA UNIV AUTONOMA DE PUEBLA on November 4, 2020 at 06:01:28 (UTC).

for ions of higher mass. In practice, high resolution designates


Mass Resolution, Mass Resolving Power 708
a mass analyzer with resolving power m/Δm50% > 10 000,
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Mass Resolution and Accuracy 708


thereby excluding quadrupole mass filter, triple quadrupole, and
Time-of-Flight Mass Analyzers 708
quadrupole ion trap mass analyzers. Finally, mass measurement
Orthogonal Acceleration (see ref 13) 709
accuracy is usually expressed as the rms difference (in ppm)
Reflectron/Multipass TOF 709
Recent Advances in TOF Mass Analyzers 709
between measured and exact (based on elemental composition)
Selected Applications 710 mass. In the absence of systematic error, mass accuracy is the
same as mass precision.


Fourier Transform Mass Analyzers 710
Common Features of Fourier Transform Mass
MASS RESOLUTION AND ACCURACY
Analyzers 710
Ion Accumulation and Detection 711 In a mass analyzer, the detected ion signal (in this case, voltage,
Advances in Fourier Transform Mass Analyzers 711 current, or time-of-flight) is digitized. Peak height and position
Selected Applications 713 are then determined by some sort of interpolation or fitting
Author Information 715 procedure.2−4 Mass measurement precision, namely, m/δm, in
Corresponding Author 715 which δm is the rms error from a large number of repeated
Biographies 715 measurements, may in general be predicted from the product of
Acknowledgments 715 signal-to-noise ratio and the square root of the number of data
References 715 points per peak width,5 depending on the nature of the noise,6
from a single spectrum. Thus, for a given mass resolution, mass


measurement precision can be increased by sampling more data
MASS RESOLUTION, MASS RESOLVING POWER points per peak width, accounting in a significant part for the
This review focuses on recent advances in instrumentation, increase in TOF mass accuracy as faster digitizers became available.
techniques, and applications of the highest resolution mass Once the peak positions have been precisely established, the
analyzers (multipass time of flight (TOF), orbitrap, and Fourier spectrum is calibrated, on the basis of the exact masses of two
transform ion cyclotron resonance (FTICR)) since a prior or more ions of known elemental composition. External
similar Analytical Chemistry review in 2008.1 We begin with calibration (performed for a different sample than for analyte)
a brief discussion of the need for high mass resolution, and is typically 2−3× less accurate than internal calibration (based
then proceed to mass analyzer types and their development on multiple ions of known m/z values in the analyte sample).
and applications for bio-analysis (e.g., proteomics, lipidomics, As the number of atoms increases, the number of possible
drug discovery) and complex organic mixtures (environmental, elemental compositions for a given experimental mass measure-
petroleum, metabolomics). ment increases.7 For proteins, the number of amino acid
Mass resolution is the minimum mass difference, m2 − m1, compositions also increases.8,9 Moreover, high resolving power is
between two mass spectral peaks such that the valley between needed to resolve closely spaced mass doublets in complex
their sum is a specified fraction (e.g., 50%) of the height of the mixtures. For example, resolution of two equally abundant ions of
smaller individual peak. For two peaks of equal height, the same nominal mass but differing in elemental composition by
m2 − m1 ≈ Δm50%, in which Δm50% is the full width at half- C3 vs SH4 (separated by ∼3.4 mDa) at m/z 700 requires a
maximum height of either peak alone. Mass resolving power is resolving power of m/Δm50% higher than 200 000.
m2/(m2 − m1), in which (m2 − m1) is mass resolution; for an
isolated peak of mass, m, mass resolving power is m/Δm50%,
and for two peaks of equal height, mass resolving power is
■ TIME-OF-FLIGHT MASS ANALYZERS
The time-of-flight mass analyzer, initially proposed by Stephan
m2/Δm50%.1 For a multiply charged ion, m is replaced in the in 194610 and reduced to practice in 1948,11 has evolved
above definitions by m/z, in which z is the ion charge in through several stages. Delayed ion extraction addressed the
multiples of the elementary charge. The ability to distinguish
ions of different elemental composition is determined by mass
Special Issue: Fundamental and Applied Reviews in Analytical
resolution (e.g., m2 − m1 = 0.0034 Da for ions differing in
Chemistry
composition by C3 vs SH4). However, mass analyzer performance
is usually expressed in terms of mass resolving power. Thus, higher Published: January 17, 2012

© 2012 American Chemical Society 708 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719
Analytical Chemistry Review

problem of kinetic energy spread among ions of the same


m/z.12 The advent of fast digitizers; matrix-assisted laser
desorption/ionization (MALDI), and MALDI imaging; orthog-
onal ion introduction; and kinetic energy focusing by use of a
reflectron brought TOF MS to its present level of popularity.
Orthogonal Acceleration (see ref 13). Ideally, a TOF
MS experiment should begin with all ions with the same initial
position and velocity. Orthogonal ion introduction, in which
ions are accelerated in a direction perpendicular to a collimated
ion beam, effectively achieves that condition and thus improves
mass resolving power. Another advantage of orthogonal
introduction is that ions can be accumulated in the acceleration
region while previously accumulated ions fly toward the detector,
thereby more efficiently enabling coupling of continuous ion
sources with TOF mass analysis. Further optimization of ion
guides and use of an ion funnel14 with automatic gain control
(AGC) improves the ion beam quality, for higher sensitivity
and mass accuracy.15
Reflectron/Multipass TOF. The resolving power for a
TOF mass analyzer is m/Δm50% = (T/2Δt), in which T is the
ion total flight time and Δt is the mass spectral peak width.
Increasing the flight time for improved resolving power is made
possible by the reflectron, introduced by Mamyrin in 1973.16
After an initial drift region, ions are subjected to a spatially
quadratic potential that acts as an ion mirror. When ions of a
given m/z but different kinetic energy fly through the reflectron
region, the faster ions penetrate farther into the reflectron (and
therefore travel a greater distance to reach the detector) than
slow ions, and ions of all speeds arrive at the detector at the
same time. The reflectron thus provides increased ion flight path, Figure 1. High resolution time-of-flight mass analyzers. Top: Dual
without defocusing due to spread in initial velocity, thereby stage reflectron. Reproduced with permission from Waters Corpo-
increasing mass resolving power. Broadband mass resolving power ration. Copyright 2009 Waters Corportation. Middle: Multipass
of 10 000 and rms mass error of 5−10 ppm may now be routinely reflectron with linear segments. Reprinted with permission from
attained. With high field pusher and dual stage reflectrons, ref 223. Bottom: Multipass spiral configuration. Reproduced with
permission from JEOL USA. Copyright 2006−2011 JEOL Ltd.
commercial TOF mass analyzers can now reach mass resolving
power of 40 00017 (Figure 1, top). If the ion spatial focusing can The most recent multipass designs include doughnut-shaped
be maintained with minimal ion loss at each reflection, the ion ion optics,23 a spiral path electric reflector24 and multiple angle
flight path may be extended by multiple reflections. Multipass reflecting electrostatic ion mirrors25 (Figure 1, middle). The
(Figure 1, middle)18 and spiral (Figure 1, bottom)19 TOF mass
latter two designs have achieved broadband mass resolving
analyzers now attain mass resolving power of 50 000 or higher.
power of 60 000.
Recently, an optimized multipass time-of-flight mass analyzer
Recent Advances in TOF Mass Analyzers. Detection. The
has been combined with MALDI for imaging mass spectrom-
etry. The multipass time-of-flight mass analyzer consists of four most commonly used TOF ion detector is the microchannel
electric sectors, with cylindrical side electrodes and Matsuda plate (MCP) detector, which converts ions to secondary electrons,
configuration to generate a toroidal electric field, in which with large active area and rapid response time. However,
focusing is maintained by quadrupole triplet lenses at the secondary electron generation efficiency varies directly with
entrance and exit of each toroidal segment. Mass resolving incident ion velocity, and ion velocity varies inversely with the
power of 130 000 has been achieved for angiotensin II square root of accelerated ion mass, so that MCP detection
[M + H]+ ions (m/z 1046.542) after 500 turns and 654.8 m efficiency for high-mass ions is low. The mechanical nano-
total flight path.20 In a typical multipass time-of-flight mass membrane detector can detect the time-varying field emission
analyzer, the power supply for ion injection/ejection is very of electrons from mechanical oscillation without mass dis-
large and complicated in order to reduce instability of voltage crimination and thus improve high-mass detection sensitivity.
switching between ion injection and ejection events. A more For example, singly charged immunoglobulin G (150 kDa) has
recent multipass TOF design adds two more sectors for ion been experimentally observed by TOF MS with nanomembrane
injection and ejection only, thereby minimizing the size of the detection.26 A different nanomechanical resonator,27,28 whose
power supply.21 resonance frequency is perturbed by surface adsorption of ions,
Although a cyclic multipass TOF mass analyzer offers was also recently reported as a sensitive mass sensor, but
potentially high mass resolution, the observable mass range is further investigation for its practical use in TOF MS is needed.
limited, because low-m/z ions eventually overtake higher-m/z When applied to TOF MS, cryogenic detectors measure low-
ions so that the effective m/z range is reduced by a factor of N energy solid-state phonon excitation created by a particle
for N passes. An elliptical flight path composed of four toroidal impact. The energy of the phonons is less than a few meV,
electric sectors works with a new segmentation method to much smaller than the energy in the electronvolt range
identify the number of laps transited by ions of a given m/z.22 needed to produce secondary electrons in a conventional
709 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719
Analytical Chemistry Review

ionization detector. The cryodetector is capable of detecting in brain areas with ischemic injury.50 As for protein profiling, a
high m/z, slow-moving ions.29 For example, a strong signal at different lipid distribution between normal and cancer cells
510 kDa from a disulfide-linked dimer has been experimentally could diagnose disease.51,52
observed in a MALDI TOF mass spectrometer coupled with a In aerosol analysis, it is necessary to track changes in aerosol
cryodetector.30 size and chemistry with subsecond time resolution. A TOF
TOF/TOF. Two time-of-flight (TOF) mass analyzers in mass analyzer collects high resolution aerosol mass spectra at
succession enable MS2 experiments.31,32 Usually, the first TOF rates exceeding 1 kHz for determination of both aerosol size
mass analyzer selects the precursor ion for injection into a high and mass.53 Alternatively, two-dimensional gas chromatography
energy collision cell for collision induced dissociation (CID). The (GCxGC) mass spectrometry requires fast scanning because
second TOF mass analyzer then records the fragment ions. The the final GC peak width is typically 20−200 ms,54 making TOF
most common TOF/TOF configuration is a linear TOF mass the MS method of choice. GCxGC TOF MS applications include
analyzer followed by a reflectron TOF mass analyzer.33,34 However, identification of naphthentic acids in oil samples,55,56 high
low resolution in MS1 can render product ion identification throughput metabolic profiling,57,58 characterization of pathogen
difficult. bacteria,59 and analysis of organic nitrogen compounds in aerosol
Multipass TOF/TOF has been achieved with a MALDI ion samples.60 Moreover, fast transient temporal analysis of catalyst
source, a multiturn TOF mass analyzer (TOF1), a collision cell, kinetics targets TOF mass analysis for its sensitivity, detector
and a quadratic-field ion mirror,35,36 providing resolving power response, and time resolution.61
of 5000 for precursor ion selection and 1000 resolving power The TOF mass analyzer has also been applied to polymer analysis
for product ions throughout the mass range. The identification by virtue of its high m/z range and MS/MS capability. Evaluation of
of lyso-phosphatidylcholine (LPC) and phosphorylation37 has top-down TOF MS/MS for poly(α-peptoid)s synthesized by N-
been demonstrated. Recently, a spiral ion optical TOF mass heterocyclic carbine (NHC)-mediated zwitterionic ring-opening
analyzer for MS1 was combined with an offset parabolic ion polymerization62 showed that electrospray ionization (ESI) enabled
reflectron TOF as MS2, providing high resolution MS1 detection of the intact molecular species, whereas MALDI resulted in
precursor monoisotopic ions up to m/z 2500.38 elimination of the NHC initiator in the presence of cationizing salts.
Selected Applications. Although TOF mass analyzers Coupled with MALDI, TOF MS can address the mechanism of
have lower mass resolution than FT mass analyzers (FTICR polymer backbone degradation via free radical chemistry63 and
and orbitrap), they have no upper m/z limit in principle and are quantify residual polyethylene glycol (PEG) in ethoxylated sur-
thus particularly useful for identifying singly charged ions of factants.64 Cation adducts generated more informative fragment
high molecular weight (as from MALDI). Fast response/scan ions under CID,65 and TOF MS has characterized various polymers
rate is also advantageous for applications requiring short acquisi- with ammonium adducts generated by ESI.66 Miscellaneous
tion period, e.g., analysis by liquid or gas chromatography mass other applications include characterization of C4 explosives by
spectrometry. TOF secondary ion mass spectrometry (SIMS),67 qualitative and
A method for screening doping agents in human urine quantitative analysis of herbal medicines (HMs) by LC/TOF
consists of solid-phase extraction followed by HPLC-TOFMS. MS,68 and identification of different contaminants in the human
Coupled with orthogonal electrospray ionization, 124 sub- body by GC/TOF MS.69
stances, including stimulants, narcotics, agonists, diuretics, etc.,
are identified in less than 30 min.39 Further development of the
extraction method enabled identification of 40 more doping
■ FOURIER TRANSFORM MASS ANALYZERS
The FTICR mass analyzer, introduced in 1974,70 has the
compounds.40 The utility of the LC-TOF method was assessed highest mass resolving power71,72 and best mass measurement
by parallel analysis of 30 authentic urine samples by use of rapid accuracy71 among current mass analyzers. The orbitrap, another
emergency drug identification and led to identification of twice Fourier transform mass analyzer, invented in 1999,73 has been
as many drugs.41 Compared to prior methods, the author widely distributed since its commercial introduction in 2004.74
concluded that UPLC-TOFMS offers an attractive alternative Common Features of Fourier Transform Mass
toxicological screening technique. Because the mass accuracy Analyzers. The ion cyclotron and orbitrap each produce a
tolerance for broad toxicology screening has historically been spatially coherent packet of ions of a given m/z. Coherence in
quite wide (20 ppm or more),42 TOF mass accuracy less than FTICR is achieved by rf excitation at the ion cyclotron
5 ppm provided substantial improvement in the number and frequency, whereas coherence in the orbitrap is achieved by
confidence of identifications.43,44 injecting ions of a given m/z into the mass analyzer in a time
Mass spectrometry imaging (MSI) provides rapid detection, short compared to the ion oscillation frequency. In both
localization, and identification of organic components of devices, the periodic motion (rotation in ICR, oscillation in
complex biological mixtures,45 to establish chemical correla- orbitrap) is detected from the oscillating current induced in
tions with biological function or morphology. MSI experiments opposed detection electrodes, as ions pass near each electrode.
are typically performed with a TOF mass analyzer, due to its The signal from ions of a given m/z is linearly proportional to
good sensitivity and speed. Localization of peptides by MALDI the number of those ions and to the radius75,76 or amplitude of
TOF imaging reveals the distribution of their respective the ion motion. Linearity is especially important for FTICR,
proteins.46 Comparison of protein profiles from normal and because ions of different m/z may be selected/excited in any
tumor tissue can lead to biomarker candidates.47,48 The desired combination by a time-domain excitation waveform
profiling and imaging of proteins involved in proliferation, produced by inverse Fourier transformation of the desired
differentiation, and apoptosis by MALDI TOF MS generates frequency-domain excitation profile (“stored waveform inverse
unique and differential proteomic blueprints for implantation Fourier transform” (SWIFT) excitation).77−79 Both devices
and interimplantation sites.49 Lipid imaging in rat brain tissues inherently detect ions of a wide m/z range simultaneously. The
during focal cerebral ischemia reveals dynamic conversion from time-domain signal is then digitized, apodized, zero-filled80,81
phosphatidylcholine (PC) to lyso-phosphatidylcholine (LPC) (to improve digital resolution), and subjected to discrete Fourier
710 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719
Analytical Chemistry Review

transformation to yield mathematically “real” and “imaginary” resolved an isotopic distribution at a resolving power in excess
frequency-domain spectra, Re(ω) and Im(ω), which are typically of 600 000 at m/z 195.90
combined to yield a “magnitude” spectrum, M(ω) (see below).76 Electric Field Optimization. The orbitrap mass analyzer is
Spectral frequency is then converted to m/z by an appropriate composed of a spindle-like central electrode and a barrel-like
calibration equation (see below). outer electrode (Figure 2). Application of a dc voltage to the
FTMS resolving power is ultimately limited by the acquisition
period, T, for the time-domain transient. However, the time-domain
transient duration is in turn limited by magnetic and electric field
imperfection, ion-neutral collisions, and ion−ion interactions.81−84
Apodization functions85 help to detect small peaks near large peaks
by reducing the magnitude of auxiliary maxima and by narrowing
the peak width near the peak base (at the cost of broadening the
peak width at half-maximum peak height).
Ion Accumulation and Detection. Both ICR and
orbitrap mass analyzers are pulsed detectors. Because ion introduc-
tion is often temporally continuous (e.g., ESI and APPI), ions
are typically accumulated externally during detection of ions
from the preceding accumulation period.86 In FTICR, ions are
externally accumulated in a multipole electric ion trap and Figure 2. Standard orbitrap (left) and compact high field orbitrap
simultaneously ejected toward the ICR cell, whereas in the orbitrap, (right) mass analyzers. Reproduced with permission of ThermoFisher
ions are collected in a “C” trap,87 and injected simultaneously Scientific, ref 90. Copyright 2009 ThermoFisher Scientific.
toward the orbitrap. In ICR, ion spatial coherence is maintained
by the confining magnetic and electrostatic fields and by ion− two axial electrodes produces an electrostatic potential that
ion interactions,84 whereas in the orbitrap, ion axial coherence ideally is the sum of a quadrupole ion trap potential and a
is achieved by short pulsed injection from the C trap and radial logarithmic potential of a cylindrical capacitor. For ICR, a
coherence is lost as ions spread out to form a rotating ring strong homogeneous static magnetic field confines ion motion
(actually, an advantage, by reducing space charge repulsions transverse to the magnetic field, and an ideally three-dimensional
and enabling increased dynamic range), and the ring for ions of quadrupolar electrostatic potential confines ion motion parallel
a given m/z oscillates axially to produce an image current on to the magnetic field.
the detection electrodes. The cyclotron frequency in ICR signal In practice, the potentials in both mass analyzers deviate
varies as (m/z)−1,75 whereas the orbitrap axial frequency (like from ideal, because the electrodes are truncated, imperfect in
the ICR “trapping” oscillation) varies as (m/z)−1/2.87 As a result, shape and alignment, and must have apertures to admit ions
ICR mass resolving power varies as (m/z)−1 vs (m/z)−1/2 for (both) and electrons or photons (ICR). In ICR, an imperfect
the orbitrap. (anharmonic) quadrupolar electrostatic potential produces an
Advances in Fourier Transform Mass Analyzers. High axially dependent, nonlinear radial electric field (Er) that makes
Field Strength. The simplest way to improve FTMS perform- the observed cyclotron frequencies depend on ion radius and
ance is to operate at higher magnetic field, B (ICR) or higher axial amplitude. Different approaches to the cell anharmonicity
electric field (orbitrap). ICR mass resolving power or scan rate problem have been investigated since the 1980s.91−99 One
increases proportional to B, whereas mass accuracy, dynamic range, approach is to insert compensation rings into an open cylindrical
and upper m/z limit increases as B2.88,89 The highest field FTICR cell100−104 (e.g., Figure 3, top). Another idea relies on ion
instrument is currently 15 T, and 21 T systems are under con- cyclotron motion to average the electric potential from curved
struction at the USA National High Magnetic Field Laboratory and segmented electrodes105 (Figure 3, bottom). Both cell designs in
Pacific Northwest National Laboratory. Figure 3 have achieved baseline unit mass resolution for proteins
The orbitrap axial oscillation frequency can be expressed up to 150 kDa.106 Moreover, a coaxial multielectrode cell
as:90 (O-trap),107 with separate compartments for ICR excitation
e 2Ur and detection, has been experimentally demonstrated. The
ω= · potential benefit from that design is enhancement of resolving
(m / z ) ⎛ R ⎞ 1
R m 2 ln⎜ 2 ⎟ − [R2 2 − R12] power equal to detected frequency multiple order that could
⎝ R1 ⎠ 2 (1) alternatively yield a shorter detection period for correspond-
−19
in which e is the elementary charge (1.602 × 10 C), R2 is the ingly higher throughput. (e.g., for LC/MS).
maximum radius of the outer barrel-like electrode, R1 is the Compared to a standard orbitrap geometry, the outer electrode
radius of the central spindle-like electrode, Ur is the voltage dimensions of the compact high-field orbitrap analyzer are scaled
applied to the central electrode, and Rm is the “characteristic” down by a factor of 1.5, whereas the central electrode dimensions
radius at which dU(r,z)/drz=0 = 0. From eq 1, it is clear that are scaled down by a factor of 1.2 (Figure 2). The entrance
orbitrap resolving power may be increased by increasing the aperture cross-section is reduced by more than a factor of 2; thus, to
central electrode voltage or decreasing the R2/R1 ratio. The avoid a corresponding loss in sensitivity, a new miniature lens system
newest orbitrap exhibits increased Ur and optimized R2/R1 ratio focuses ions to a much smaller spot. The capacitance of the orbitrap
to provide ∼2-fold improvement in resolving power (for a drops in proportion to size, which results in improved image current
given data acquisition period) or similar reduction in data detection sensitivity. New transistors in the preamplifier further
acquisition period (for faster online LC sampling, for a given increase sensitivity. In spite of higher ion density, the relatively thicker
mass resolving power). With careful balancing of construction central electrode results in smaller space charge shifts than for the
tolerances and experimental parameters, the new orbitrap standard orbitrap.
711 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719
Analytical Chemistry Review

A(ω) = cos[ϕ(ω)]Re(ω) − sin[ϕ(ω)]Im(ω) (3a)

D(ω) = sin[ϕ(ω)]Re(ω) + cos[ϕ(ω)]Im(ω) (3b)

An absorption-mode spectral peak is inherently narrower


than its corresponding magnitude-mode spectral peak (Figure 4,

Figure 3. Top: Seven segment compensated ICR cell. Repro-


duced with permission from ref 102. Copyright 2011 American
Chemical Society. Bottom: Dynamically harmonized ICR cell
(bottom). Adapted with permission from ref 105. Copyright 2010
John Wiley & Sons, Inc.

Software Development. Calibration. FTMS mass accuracy


is determined by signal-to-noise ratio and number of data
points per peak width 5 and by “calibration”, namely, the
conversion of a frequency-domain spectrum to an m/z
spectrum. For FTICR, calibration typically relies on a two-
term equation based on a perfectly homogeneous magnetic
field and a purely quadrupolar electrostatic field.108,109 Other
equations incorporating variation in ion abundance110 and space-
charge effects111,112 have also been evaluated. External calibra- Figure 4. (a) Magnitude-mode and absorption-mode Lorentzian
tion (performed for a different sample than for analyte) is spectra, corresponding to Fourier transformation of an infinite duration
typically 2−3× less accurate than internal calibration (based on time-domain signal, f(t) = A cos(ω0t) exp(−t/τ). (b) Magnitude (upper)
multiple ions of known m/z values in the analyte sample). and absorption-mode (lower) electrospray ionization 9.4 T
Systematic error has been reduced by a factor of up to 3 (and FTICR mass spectra from the same bitumen time-domain data.
the number of assigned peaks increased by ∼25%) by separate The absorption display clearly resolves a mass doublet (com-
calibration for each of up to ∼30 individual mass spectral positions differing by C3 vs SH4, 0.0034 Da) unresolved in magnitude
segments.113,71 Addition of an ion abundance-dependent term mode.
to the calibration equation can reduce systematic error by
another factor of ∼2−3.71,103 For a high vacuum gas oil sample top) by a factor that depends on the presence and mechanism of
with ∼5200 peaks, FTICR rms mass error for an absorption- signal damping,81−84 for an improvement in mass resolving power
mode spectrum (see below) drops from 107 to 27 ppb by by a factor of up to 2. In FTICR, temporally dispersed excitation
essentially eliminating systematic error. For the orbitrap, m/z is and the delay between excitation and detection result in complex
first-order proportional to (ω)−1/2, leading to a one term variation of phase with frequency. For that reason, magnitude
calibration equation. As for ICR, orbitrap mass calibration may mode has been the conventional display for FTICR mass spectra.
be improved by considering the effect of space charge, leading The advantage of absorption-mode display was recognized from
to a mass measurement error as low as ∼80 ppb over an m/z the outset.115 Efforts to achieve broadband-phase correction116,117
range of 100−2000.114 culminated in a phase correction algorithm that can automatically
Phase Correction. Fourier transformation of a time-domain account for first- and second-order-phase accumulation from
signal produces mathematically real and imaginary frequency- excitation parameters and iterates the zero-order term.118 An
domain spectra, Re(ω) and Im(ω), which may be combined to experimental FTICR absorption-mode mass spectrum with 40%
yield magnitude-mode (also known as absolute-value) and phase higher resolving power (and thus an increased number of resolved
spectra, M(ω) and φ(ω), related according to eqs 2a and 2b.76 peaks) as well as higher mass accuracy relative to its corresponding
magnitude mode spectrum is shown in Figure 4, bottom. Another
M(ω) = [[Re(ω)]2 + [Im(ω)]2 ]1/2 (2a) approach for phase correction is to assume a quadratic relation
between phase and frequency and iteratively solve for zero-, first-,
ϕ(ω) = arc tan[Im(ω)/Re(ω)] (2b)
and second-order coefficients.119
If the “phase spectrum”, φ(ω), is known, absorption- and ICR phase “wrapping” (i.e., phase variation by more than 2π
dispersion-mode spectra, A(ω) and D(ω), may in turn be across the frequency spectrum) for the orbitrap is less severe
obtained as linear combinations of Re(ω) and Im(ω). due to the short ion injection period from the C trap but
712 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719
Analytical Chemistry Review

complicated by the change in central electrode voltage (and post-translational modification according to its characteristic addi-
therefore resonant frequency) during ion injection. A “pseudo”- tional mass relative to an unmodified amino acid residue. The top-
phase correction for the orbitrap combines absorption-mode down approach can in principle locate all post-translational modifi-
display for the top part of the peak with an increasing magnitude- cations, even when several are present at the same time. High
mode component for the bottom part. Although the peak width resolution and high mass accuracy are essential to resolve isotopic
at half peak height is thus narrower,120 improvement vanishes distributions from peptides of different charge state as well as to
for low abundance peaks that are near the base of a high assign isobaric peptide compositions (e.g., lysine vs glutamine,
abundance peak, and the mass measurement accuracy does differing in mass by 0.0364 Da).129 Examples of top-down post-
not improve. translational identifications include: rhesus monkey cardiac
Improved Data Station. Continued improvement in FTz troponin,130 integral membrane proteins,131 and human histo-
mass analyzers requires optimized control of multiple nes.132,133 Further, 99 proteins have been identified by top-down
experimental events, precisely timed data acquisition, and efficient tandem mass spectrometry in Methanosarcina acetivorans,134
data analysis to accommodate new hardware modifications and and capillary liquid chromatography coupled with FTICR
ion optics configurations. A recent Predator data station121 identified with high confidence 102 endogenous peptides in the
exploits improved PC bus speed to transfer data to and from suprachiasmatic nucleus. Robust two-dimensional liquid
the control PC in real time. Conditional data acquisition can chromatography combined with top down mass spectrometry
evaluate data in real time and apply user-defined conditions to increases efficiency for improved quantification and character-
average and store data. This capability is primarily used to eliminate ization.135,136 Further automated data processing can increase
low quality data so as to improve the data S/N ratio and top-down throughput.137 In addition to conventional ESI, IR-
minimize the computer memory required to store data. Tool MALDESI has been coupled with an FTICR mass analyzer to
command language (Tcl) scripting has been applied for 17 perform top-down analysis of equine myoglobin (17 kDa).138
voltages and 18 triggers and provides a fast and easy way to Discovery and characterization of endogenous peptide139 and
control the Predator data station and associated peripherals, candidate pharmacodynamic markers140 has been demonstrated.
e.g., LC, autosampler, laser source, and electron emitter.121 Another Recently, FTICR MS achieved unit mass baseline resolution for
improved data control system combines PXI hardware with a an intact 148 kDa therapeutic monoclonal antibody, the highest
workflow-based acquisition and control software to allow unique mass protein isotopically resolved to date.106
types of data-dependent experiments.122 Dynamic switching In the “bottom-up” approach, proteins are typically separated
between different dissociation technologies may be achieved due by HPLC and/or electrophoresis, proteolytically digested into
to fast and precise hardware response.123 peptides, and then subjected to online LC/MS, and the most
Selected Applications. Tandem Mass Spectrometry. abundant peptides are subjected to MS/MS.141,142 Any of
The main difference between ICR and orbitrap for MS/MS is several algorithms143,144 then compare precursor ion mass and
that precursor ion dissociation can be performed in a Penning MS/MS peak spacing to an appropriate database for protein
trap but not in an orbitrap. In principle, fragment ions can be identification.145−147 Identification reliability may be improved
brought to coherence in an orbitrap for subsequent excitation by incorporating LC elution time.148 Alternatively, “shotgun”
and detection,124 but MS/MS is typically performed externally proteomics also relies on online LC/MS and MS/MS but
(CID or ETD) for orbitrap detection.125,126 MS/MS can be without prior gel separation of the proteins.149 Shotgun
performed in an ICR cell by collision-induced dissociation proteomics has been applied to microorganisms,150 histones
(CID), infrared multiphoton dissociation (IRMPD), and (especially post-translational modifications),151,152 and discov-
electron capture dissociation (ECD). However, high resolution ery of biosynthetic pathways,153 etc. For proteins too large for
requires low pressure in the ICR cell, so introduction of collision efficient gas-phase fragmentation, “middle-down” proteomics
gas into the cell requires subsequent pumpdown before denotes LC/MS and MS/MS for large proteolyic fragments of
excitation/detection; ergo, CID FTICR experiments are now the original protein and has identified 7454 peptides from 2 to
typically conducted with an external electric multipole ion trap. 20 kDa after 23 LC MS/MS injections of Lys-C digests of
Because ECD (or ETD) can fragment a peptide but not dissociate HeLa-S3 nuclear proteins.154 Post-translationally modified
the noncovalently bound fragments, infrared irradiation is histone H3 variants have also been characterized by that
commonly used to release the product ion. CID and IRMPD approach.155
heat an ion and typically break the weakest bond to produce Quantitative proteomics typically requires isotopic labeling,
predominantly b- and z-type peptide fragment ions. However, either before (SILAC)156 or after peptide isolation.142 Quantifica-
post-translational linkages (e.g., phosphorylation, glycosylation) tion of human monkeypox virus and vaccinia virus confirmed the
are often lost before peptide backbone cleavage, thereby eliminat- level required for pathogenesis with the accurate mass and time tag
ing knowledge of their location. In contrast, ECD (or ETD) (AMT) method after peptide isolation.157 With stable isotope
produces mainly c- and z-type backbone cleavage fragment ions by labeling of amino acids in cell culture (SILAC), it is possible to
breaking the backbone N−Cα bond without loss of phosphate or quantitatively evaluate phosphorylation changes in stable cell
glycan. Thus, ECD or ETD is much preferred for locating post- lines.158 Quantitative proteomics depends on fragmentation
translational modifications.127,128 energy.159,160 When isotopic labeling is not applicable, label-free
Proteomics. Proteomics encompasses identification and methods can provide a measure of quantitation.161
structural characterization of proteins and their complexes, Metabolomics. High throughput screening of chemical
determination of post-translational modifications, and quantitation toxicity in Daphnia magna has been reported.162 Metabolic
of protein expression under different treatments. In “top-down” profiling facilitates biochemical phenotyping of normal and
proteomics, a single gas-phase protein is isolated and subjected to neoplastic colon tissue at high significance levels163 and
fragmentation (e.g., CID, IRMPD, ECD, ETD, etc.) to generate annotates the ions originating from identical metabolites by
fragment masses for comparison to an appropriate database for fragmentation pattern and database searching analysis.164 Coupled
protein identification, as well as identity and location of each with different chromatography platforms,165−167 higher dynamic
713 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719
Analytical Chemistry Review

range and identification of more metabolites can be achieved. ligands,184 enable characterization of polypurine tract-containing
Autocorrelation of retention and ionization leads to more RNA−DNA hybrids and show that the common structural
exhaustive metabolic fingerprints.168 features of lentiviral and retrotransposon polypurine tracts
Lipidomics. Characterization of nonpolar lipids and selected facilitate the interaction with their cognate reverse transcriptase.
steroids by high resolution mass analysis has emerged as a Structural elucidation of the HIV-1 virus has also been achieved
powerful tool for lipidomics.169,170 Online LC separation can by the combination of footprinting and cross-linking techniques
quickly narrow down the possible phospholipid and glyco- with high resolution MS detection.184,185
sphingolipid compositions and facilitate their identifica- Synthetic Polymer Analysis. The most obvious advantage of
tion.171,172 Unequivocal lipid elemental composition from mass spectrometry for synthetic polymer analysis is the generation
isotopic fine structure has been used to validate the of the full molecular weight distribution, rather than simply
identification of sulfur-containing lipids in algae extracts number-average or weight-average molecular weight.186,187 High
(Figure 5).173 Direct infusion high resolution mass analysis with resolution MS can also reveal pathways of polymer synthesis, as
for a sterically hindered alkoxyamine initiator for the nitroxide-
mediated copolymerization (NMP) of methyl methacrylate
(MMA);188 the styrene monomer was added as polymerization
terminator, and the progress was monitored from the product
oligomer distributions. Synthetic polymer structure has been
probed by various fragmentation techniques, including electron
capture dissociation (ECD), collision induced dissociation (CID),
and electron detachment dissociation (EDD),189−194 e.g.,
polyamidoamine dendrimer characterization by ECD and EDD.
Petroleomics. From sufficiently complete characterization
of the organic composition of petroleum and its products, it is
becoming possible to correlate (and ultimately predict) their
properties and behavior.195,196 FTICR MS is the preferred
identification technique for petroleum analysis because of the
need to resolve mass splits down to 3.4 mDa (32S1H4 vs 12C3) or
∼0.5 mDa (for nickel-containing porphyrins) (Figure 6). Removal
Figure 5. Positive electrospray ionization 14.5 T FTICR mass
spectrum for Nannochloropsis oculata species lipid extract. Inset: Two
peaks differing by 2.37 mDa are resolved and assigned. (Figure kindly
provided by Huan He.)

computer-assisted assignment is able to profile the


13
C-isotopologues of glycerophospholipids (GPL) directly in
crude cell extracts.174 High resolution MS/MS enabled
identification of triacylglycerols archeological extracts175 and
neutral lipids.176 Matrix-assisted laser desorption ionization
combined with high-resolution MS enables confident identi-
fication and spatial localization of lipids in tissue sections.177,178
RNA/DNA Analysis. Most high resolution MS analyses of
oligonucleotides have focused on synthetic DNA, RNA,
aptamers, etc. Double-stranded small interfering RNA (siRNA)
presents numerous fragment ions and multiple chemical
modifications. The different metabolite patterns for synthetic
siRNA in different biomatrixes (rat and human serum) provide Figure 6. Positive ion atmospheric pressure photoionization (APPI)
insight into the stability and pharmacokinetic properties of 9.4 T Fourier transform ion cyclotron resonance (FTICR) mass
therapeutic siRNA compounds.179 A method for sequencing spectrum of a petroleum crude oil. Upper inset: Mass scale expansion
single and double stranded RNA oligonucleotides by acid revealing 90 singly charged ions within a mass range of 0.32 Da. Lower
hydrolysis has been presented. From the mass differences inset: Baseline resolution of ions differing in mass by 1.1 mDa, made
between adjacent members of a mass ladder, the complete possible by ultrahigh mass resolving power of 1 000 000 at m/z 428.
(Data kindly provided by Amy McKenna.)
sequences of different siRNA 21-mer single and double strands
could be verified. This simple and fast method can be applied to
of contaminants in asphaltenes during the crude oil processing is
controlling sequences of synthetic oligomers, as well as for de
necessary to avoid catalyst fouling and hence lower liquid yield.
novo sequencing.180 A high resolution mass spectrometry-based
analytical platform for RNA, employing direct nanoflow Also, asphaltenes can precipitate during production and refining.
reversed-phase liquid chromatography with a spray tip column Thus, asphaltenes are one of the most discussed topics in
predicted the nucleotide composition of a 21-nucleotide small petroleomics.198−202 Ion mobility mass spectrometry and FTICR
interfering RNA, detected yeast tRNA post-transcriptional MS have been combined to confirm the presence of asphaltene
modifications, and analyzed the nucleolytic fragments of RNA nanoaggregates in toluene at concentrations (10−4 mass fraction)
at a subfemtomole level.181 Various other approaches, e.g., below those in crude oils.197 Subsequently, in situ analysis of a
combing chemical modification of RNA with mass spectrom- 3000 ft vertical column of crude oil by downhole fluid analysis
etry,182 based on neomycin B+183 or classical nucleic acid indicated that the asphaltenes in a black crude oil exhibit
714 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719
Analytical Chemistry Review

gravitational sedimentation according to the Boltzmann distribu- development, and applications of analytical Fourier transform ion
tion and that the asphaltene colloidal size is ∼2 nm.203 The cyclotron resonance mass spectrometry.
detailed characterization of a Middle Eastern heavy crude oil Feng Xian is a PhD candidate in the Ion Cyclotron Resonance
distillation series fully validated the Boduszynski model, namely, program at the National High Magnetic Field Laboratory at Florida
that petroleum is a continuum with regard to composition, State University in Tallahassee, Florida. He received his Bachelor of
molecular weight, aromaticity, and heteroatom content and that Science in Chemical Engineering from the Xi’an Jiaotong University in
boiling point can be predicted from aromaticity and heteroatom 1996, and he anticipates his Ph.D. in Analytical Chemistry from Florida
content.204,205 Naphthenic acids (i.e., species containing non- State University in 2012. His research focuses on Fourier Transform
aromatic hydrocarbon rings) can form harmful deposits and may Ion Cyclotron Resonance (FTICR) mass spectrometry instrumenta-
be identified in crude oil before the deposits form206−208 as well as tion development.
in oil sands processed water.209−211 Ultrahigh mass resolution also
enables identification of biomarkers, such as nickel and vanadyl
petroporphyrins.212,213
Environmental Analysis. Applications of high resolution
■ ACKNOWLEDGMENTS
The authors are not aware of any biases that might be perceived
MS technology in the fields of food safety214 and environmental as affecting the objectivity of this Review. We thank Amy
analysis have recently been reviewed.215 High resolution helps to McKenna and Huan He for providing the data for Figures 5
trace contaminants, as well as to identify analogs for which and 6, and Alexander Makarov for providing images for Figure 2.
standards are not available. High-resolution full scan MS analysis
This work was supported by NSF Division of Materials
with simultaneous targeted CID MS/MS has been applied to
Research through DMR-0654118 and the State of Florida.
analysis of polyether toxin azaspiracid food contaminants in
shellfish.214 High resolution is also useful in the high-throughput
screening of micropollutants in wastewater treatment. Both
targeted and nontargeted screening have identified a variety of
■ REFERENCES
(1) Marshall, A. G.; Hendrickson, C. L. Ann. Rev. Anal. Chem. 2008,
unreported and previously reported pesticide transformation 1, 579−599.
products.216 Characteristic polar oil sand components (naphthenic (2) Giancaspro, C.; Comisarow, M. B. Appl. Spectrosc. 1983, 37, 153−
acids and other related acid fraction components) spiked into plant 166.
tissue prior to extraction can be differentiated from coextracted (3) Keefe, C. D.; Comisarow, M. B. Appl. Spectrosc. 1989, 43, 605−
endogenous plant components by high resolution MS.210 LC/high 607.
resolution MS showed that the industrial chemical, perfluoroocta- (4) Verdun, F. R.; Giancaspro, C.; Marshall, A. G. Appl. Spectrosc.
noic acid, is microbiologically inert and hence environmentally 1988, 42, 715−721.
(5) Chen, L.; Cottrell, C. E.; Marshall, A. G. Chemom. Intell. Lab. Syst.
persistent.217 The high production volume chemicals, benzotria-
1986, 1, 51−58.
zoles and benzothiazoles, have been extracted from environmental (6) Liang, Z.; Marshall, A. G. Appl. Spectrosc. 1990, 44, 766−775.
water by solid-phase extraction and identified by LC/high (7) Kim, S.; Rodgers, R. P.; Marshall, A. G. Int. J. Mass Spectrom.
resolution MS.218 Induced metabolite changes in myriophyllum 2006, 251, 260−265.
spicatum, an aquatic vascular plant, have been monitored by high (8) He, F.; Emmett, M. R.; Hakansson, K.; Hendrickson, C. L.;
resolution MS.219 The method has also provided molecular Marshall, A. G. J. Proteome Res. 2004, 3, 61−67.
characterization as the level of elemental composition, for dissolved (9) Mao, Y.; Tipton, J. D.; Blakney, G. T.; Hendrickson, C. L.;
organic matter (DOM), e.g., from pore water,220 secondary-treated Marshall, A. G. Anal. Chem. 2011, 83, 8024−8028.
wastewater,221 and the Greenland ice sheet,222 because it is capable (10) Mirsaleh-Kohan, N.; Robertson, W. D.; Compton, R. N. Mass
of resolving complex molecular mixtures and providing information Spectrom. Rev. 2008, 27, 237−285.
about the exact elemental composition. (11) Cameron, A. E.; Eggers, D. F. Rev. Sci. Instrum. 1948, 19, 605−


607.
AUTHOR INFORMATION (12) Wiley, W. C.; McLaren, I. H. Rev. Sci. Instrum. 1955, 26, 1150−
1157.
Corresponding Author (13) Verentchikov, A. N.; Ens, W.; Standing, K. G. Anal. Chem. 1994,
*E-mail: marshall@magnet.fsu.edu (A.G.M.); hendrick@ 66, 126−133.
magnet.fsu.edu (C.L.H.). (14) Kelly, R. T.; Tolmachev, A. V.; Page, J. S.; Tang, K.; Smith, R. D.
Biographies Mass Spectrom. Rev. 2010, 29, 294−312.
(15) Ibrahim, Y. M.; Belov, M. E.; Liyu, A. V.; Smith, R. D. Anal.
Alan G. Marshall completed his B.A. degree with Honors in Chem. 2008, 80, 5367−5376.
Chemistry at Northwestern University in 1965 and his Ph.D. in (16) Mamyrin, B. A. Int. J. Mass Spectrom. 2000, 206, 251−266.
Physical Chemistry from Stanford University in 1970. He is Robert O. (17) Pringle, S. D.; Giles, K.; Wildgoose, J. L.; Williams, J. P.; Slade,
Lawton Professor of Chemistry and Biochemistry at Florida State S. E.; Thalassinos, K.; Bateman, R. H.; Bowers, M. T.; Scrivens, J. H.
University and Director of the Ion Cyclotron Resonance (ICR) Int. J. Mass Spectrom. 2007, 261, 1−12.
Program, an NSF national user facility for mass spectrometry. His (18) Toyoda, M.; Okumura, D.; Ishihara, M.; Katakuse, I. J. Mass
current reseach spans FTICR instrumentation development, fossil Spectrom. 2003, 38, 1125−1142.
fuels and environmental analysis, and mapping the primary and higher- (19) Satoh, T.; Sato, T.; Tamura, J. J. Am. Soc. Mass Spectrom. 2007,
order structures of biological macromolecules and their complexes. 18, 1318−1323.
(20) Hazama, H.; Aoki, J.; Nagao, H.; Suzuki, R.; Tashima, T.; Fujii,
Christopher L. Hendrickson, Associate Director of the Ion Cyclotron K.-i.; Masuda, K.; Awazu, K.; Toyoda, M.; Naito, Y. Appl. Surf. Sci.
Resonance Program at the National High Magnetic Field Laboratory 2008, 255, 1257−1263.
and Courtesy Professor of Chemistry at Florida State University, (21) Shimma, S.; Nagao, H.; Aoki, J.; Takahashi, K.; Miki, S.;
received his B.A. degree in chemistry from the University of Northern Toyoda, M. Anal. Chem. 2010, 82, 8456−8463.
Iowa and his Ph.D. in analytical chemistry from the University of Texas (22) Nishiguchi, M.; Ueno, Y.; Toyoda, M.; Setou, M. J. Mass
at Austin. His research interests are in instrumentation, technique Spectrom. 2008, 44, 594−604.

715 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719


Analytical Chemistry Review

(23) Shchepunov, V.; Berdnikov, A.; Lzumi, H.; Giles, R.; Gall, N. (55) Rowland, S. J.; Scarlett, A. G.; Jones, D.; West, C. E.; Frank, R. A.
Proceedings of the 58th ASMS Conference on Mass Spectrometry & Allied Environ. Sci. Technol. 2011, 45, 3154−3159.
Topics, Salt Lake City, UT, May 23−27, 2010. (56) Rowland, S. J.; West, C. E.; Scarlett, A. G.; Jones, D.; Frank, R. A.
(24) Ioanoviciu, D. Int. J. Mass Spectrom. 2010, 290, 145−147. Rapid Commun. Mass Spectrom. 2011, 25, 1198−1204.
(25) Verentchikov, A. N.; Yavor, M. I. Quasi-Planar Multi-Reflecting (57) Li, X.; Xu, Z.; Lu, X.; Ying, B.; Kong, H.; Yu, Y.; Xu, G. Anal.
Time-of-Flight Mass Spectrometer. United States Patent, US 2011/ Chim. Acta 2009, 633, 257−262.
01086729 A1, 2011 (58) Wojtowicz, P.; Zrostlikova, J.; Kovalczuk, T.; Schurek, J.; Adam, T.
(26) Park, J.; Qin, H.; Scalf, M.; Hilger, R. T.; Westphall, M. S.; J. Chromatogr., A 2010, 1217, 8054−8061.
Smith, L. M.; Blick, R. H. Nano Lett. 2011, 11, 3681−3684. (59) Gardner, J. Y.; Brillhart, D. E.; Benjamin, M. M.; Dixon, L. G.;
(27) Chiu, H.-Y.; Hung, P.; Postma, H. W. C.; Bockrath, M. Nano Mitchell, L. M.; Dimandja, J.-M. D. J. Sep. Sci. 2011, 34, 176−185.
Lett. 2008, 8, 4342−4346. (60) Ozel, M. Z.; Ward, M. W.; Hamilton, J. F.; Lewis, A. C.;
(28) Lassagne, B.; Garcia-Sanchez, D.; Aguasca, A.; Bachtold, A. Raventos-Duran, T.; Harrison, R. M. Aerosol Sci. Technol. 2009, 44,
Nano Lett. 2008, 8, 3735−3738. 109−116.
(29) Frank, M.; Labov, S. E.; Westmacott, G.; Benner, W. H. Mass (61) Goguet, A.; Hardacre, C.; Maguire, N.; Morgan, K.; Shekhtman,
Spectrom. Rev. 1999, 18, 155−186. S. O.; Thompson, S. P. Analyst 2011, 136, 155−163.
(30) Wenzel, R. J.; Matter, U.; Schultheis, L.; Zenobi, R. Anal. Chem. (62) Li, X.; Casiano-Maldonado, M.; Zhang, D.; Wesdemiotis, C.
2005, 77, 4329−4337. Macromolecules 2011, 44, 4555−4564.
(31) Cotter, R. J.; Gardner, B. D.; Iltchenko, S.; English, R. D. Anal. (63) Polce, M. J.; Ocampo, M.; Quirk, R. P.; Wesdemiotis, C. Anal.
Chem. 2004, 76, 1976−1981. Chem. 2008, 80, 347−354.
(32) Iltchenko, S.; Cotter, R. J. Int. J. Mass Spectrom. 2007, 265, 372− (64) Szyszke, R.; Hanton, S. D.; Henning, D.; Owens, K. G. J. Am.
381. Soc. Mass Spectrom. 2011, 22, 633−640.
(33) Pittenauer, E.; Allmaier, G. J. Am. Soc. Mass Spectrom. 2009, 20, (65) Nasioudis, A.; van Velde, J. W.; Heeren, R. M. A.; van den Brink,
1037−1047. O. F. Int. J. Mass Spectrom. 2011, 303, 63−68.
(34) Pittenauer, E.; Allmaier, G. Comb. Chem. High Throughput (66) Fouquet, T.; Humbel, S.; Charles, L. J. Am. Soc. Mass Spectrom.
Screening 2009, 12, 137−155. 2011, 22, 649−658.
(35) Toyoda, M. Eur. J. Mass Spectrom. 2010, 16, 397−406. (67) Mahoney, C. M.; Fahey, A. J.; Steffens, K. L.; Benner, B. A.;
(36) Toyoda, M.; Giannakopulos, A.; Colburn, A.; Derrick, P. Rev. Lareau, R. T. Anal. Chem. 2010, 82, 7237−7248.
Sci. Instrum. 2007, 78, 074101−074109. (68) Zhou, J.-L.; Qi, L.-W.; Li, P. J. Chromatogr., A 2009, 1216,
(37) Shimma, S.; Nagao, H.; Giannakopulos, A.; Hayakawa, S.; 7582−7594.
Awazu, K.; Toyoda, M. J. Mass Spectrom. 2008, 43, 535−537. (69) Hernandez, F.; Portoles, T.; Pitarch, E.; Lopez, F. J. J. Mass
(38) Satoh, T.; Sato, T.; Kubo, A.; Tamura, J. J. Am. Soc. Mass Spectrom. 2008, 44, 1−11.
Spectrom. 2011, 22, 797−803. (70) Comisarow, M. B.; Marshall, A. G. Chem. Phys. Lett. 1974, 25,
(39) Kolmonen, M.; Leinonen, A.; Pelander, A.; Ojanpera, I. Anal. 282−283.
Chim. Acta 2007, 585, 94−102. (71) Savory, J. J.; Kaiser, N. K.; McKenna, A. M.; Xian, F.; Blakney,
(40) Kolmonen, M.; Leinonen, A.; Pelander, A.; Ojanpera, I. Drug G. T.; Rodgers, R. P.; Hendrickson, C. L.; Marshall, A. G. Anal. Chem.
Test. Anal. 2009, 1, 250−266. 2011, 83, 1732−1736.
(41) Lee, H. K.; Ho, C. S.; Iu, Y. P. H; Lai, P. S. J.; Shek, C. C.; Lo, (72) Shi, S. D.-H.; Hendrickson, C. L.; Marshall, A. G. Proc. Natl.
Y.-C.; Klinke, H. B.; Wood, M. Anal. Chim. Acta 2009, 649, 80−90. Acad. Sci. U.S.A. 1998, 95, 11532−11537.
(42) Williamson, L. N.; Zhang, G. D.; Terry, A. V.; Bartlett, M. G. (73) Makarov, A. Anal. Chem. 2000, 72, 1156−1162.
J. Liq. Chromatogr. Relat. Technol. 2008, 31, 2737−2751. (74) Hu, Q.; Noll, R. J.; Li, H.; Makarov, A.; Hardman, M.; Cooks, R. G.
(43) Badoud, F.; Grate, E.; Perrenoud, L.; Avois, L.; Saugy, M.; J. Mass Spectrom. 2005, 40, 430−443.
Rudaz, S.; Venthey, J.-L. J. Chromatogr., A 2009, 1216, 4423−4433. (75) Marshall, A. G.; Hendrickson, C. L.; Jackson, G. S. Mass Spectrom.
(44) van der Heeft, E.; Bolck, Y. J. C.; Beurner, B.; Nijrolder, A. W. J. M.; Rev. 1998, 17, 1−35.
Stolker, A. A. M.; Nielen, M. W. F. J. Am. Soc. Mass Spectrom. 2009, 20, (76) Marshall, A. G.; Verdun, F. R.Fourier Transforms in NMR, Optical,
451−463. and Mass Spectrometry: A User’s Handbook; Elsevier: Amsterdam, 1990;
(45) Chughtai, K.; Heeren, R. M. A. Chem. Rev. 2010, 110, 3237− pp 460.
3277. (77) Chen, L.; Wang, T.-C. L.; Ricca, T. L.; Marshall, A. G. Anal.
(46) Tucker, K. R.; Serebryannyy, L. A.; Zimmerman, T. A.; Rubakhin, Chem. 1987, 59, 449−454.
S. S.; Sweedler, J. V. Chem. Sci. 2011, 2, 785−795. (78) Guan, S.; Marshall, A. G. Int. J. Mass Spectrom. Ion Processes
(47) Kang, S.; Shim, H. S.; Lee, J. S.; Kim, D. S.; Kim, H. Y.; Hong, 1996, 157/158, 5−37.
S. H.; Kim, P. S.; Yoon, J. H.; Cho, N. H. J. Proteome Res. 2010, 9, (79) Marshall, A. G.; Wang, T.-C. L.; Ricca, T. L. J. Am. Chem. Soc.
1157−1164. 1985, 107, 7893−7897.
(48) Sanders, M. E.; Dias, E. C.; Xu, B. J.; Mobley, J. A.; Billheimer, (80) Bartholdi, E.; Ernst, R. R. J. Magn. Reson. 1969, 1973 (11), 9−19.
D.; Roder, H.; Grigorieva, J.; Dowsett, M.; Arteaga, C. L.; Caprioli, R. M. (81) Comisarow, M. B.; Marshall, A. G. J. Chem. Phys. 1976, 64,
J. Proteome Res. 2008, 7, 1500−1506. 110−119.
(49) Burnum, K. E.; Tranguch, S.; Mi, D.; Daikoku, T.; Dey, S. K.; (82) Guan, S.; Li, G.-Z.; Marshall, A. G. Int. J. Mass Spectrom. Ion
Caprioli, R. M. Endocrinology 2008, 147, 3274−3278. Processes 1997, 167/168, 185−194.
(50) Koizumi, S.; Yamamoto, S.; Hayasaka, T.; Konishi, Y.; (83) Mitchell, D. W. J. Am. Soc. Mass Spectrom. 1999, 10, 136−152.
Yamaguchi-Okada, M.; Goto-Inoue, N.; Sugiura, Y.; Setou, M.; (84) Nikolaev, E. N.; Heeren, R. M. A.; Popov, A. M.; Pozdneev,
Namba, H. Neuroscience 2010, 168, 219−225. A. V.; Chingin, K. S. Rapid Commun. Mass Spectrom. 2007, 21, 3527−
(51) Dill, A. L.; Ifa, D. R.; Manicke, N. E.; Costa, A. B.; Ramos-Vara, 3546.
J. A.; Knapp, D. W.; Cooks, R. G. Anal. Chem. 2009, 81, 8758−8764. (85) Aarstol, M.; Comisarow, M. B. Int. J. Mass Spectrom. Ion Processes
(52) Girod, M.; Shi, Y.; Cheng, J.-X.; Cooks, R. G. J. Am. Soc. Mass 1987, 76, 287−297.
Spectrom. 2010, 21, 1177−1189. (86) Senko, M. W.; Hendrickson, C. L.; Emmett, M. R.; Shi, S. D.-H.;
(53) Kimmel, J. R.; Farmer, D. K.; Cubison, M. J.; Sueper, D.; Marshall, A. G. J. Am. Soc. Mass Spectrom. 1997, 8, 970−976.
Tanner, C.; Nemitz, E.; Worsnop, D. R.; Gonin, M.; Jimenez, J. L. Int. (87) Perry, R. H.; Cooks, R. G.; Noll, R. J. Mass Spectrom. Rev. 2008,
J. Mass Spectrom. 2011, 303, 15−26. 27, 661−699.
(54) Shellie, R.; Marriott, P. J. Flavour and Fragrance J. 2003, 18, (88) Marshall, A. G.; Guan, S. Rapid Commun. Mass Spectrom. 1996,
179−191. 10, 1819−1823.

716 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719


Analytical Chemistry Review

(89) Schaub, T. M.; Hendrickson, C. L.; Horning, S.; Quinn, J. P.; (122) Mize, T. H.; Taban, I. M.; Duursma, M.; Seynen, M.;
Senko, M. W.; Marshall, A. G. Anal. Chem. 2008, 80, 3985−3990. Konijnenburg, M.; Vijftigschild, A.; Doornik, C. V.; Rooij, G. V.;
(90) Makarov, A.; Denisov, E.; Lange, O. J. Am. Soc. Mass Spectrom. Heeren, R. M. A. Int. J. Mass Spectrom. 2004, 235, 243−253.
2009, 20, 1391−1396. (123) Taban, I. M.; van der Burgt, Y. E. M.; Duursma, M.; Takats, Z.;
(91) Bruce, J. E.; Anderson, G. A.; Lin, C. Y.; Gorshkov, M.; Seynen, M.; Konijnenburg, M.; Vijftigschild, A.; Attema, I.; Heeren, R.
Rockwood, A. L.; Smith, R. D. J. Mass Spectrom. 2000, 35, 85−94. M. A. Rapid Commun. Mass Spectrom. 2008, 22, 1245−1256.
(92) Gabrielse, G.; Haarsma, L.; Rolston, S. L. Int. J. Mass Spectrom. (124) Perry, R. H.; Hu, Q.; Salazar, G. A.; Cooks, R. G.; Noll, R. J.
1989, 88, 319−332. J. Am. Soc. Mass Spectrom. 2009, 20, 1397−1404.
(93) Gabrielse, G.; MacKintosh, F. Int. J. Mass Spectrom. Ion Processes (125) Frese, C. K.; Altelaar, A. F. M.; Hennrich, M. L.; Nolting, D.;
1984, 57, 1−17. Zeller, M.; Griep-Raming, J.; Heck, A. J. R.; Mohammed, S. J. Proteome
(94) Gooden, J. K.; Rempel, D. L.; Gross, M. L. J. Am. Soc. Mass Res. 2011, 10, 2377−2388.
Spectrom. 2004, 15, 1109−1115. (126) Macek, B.; Waanders, L. F.; Olsen, J. V.; Mann, M. Mol. Cell.
(95) Guan, S.; Marshall, A. G. Int. J. Mass Spectrom. Ion Processes Proteomics 2006, 5, 949−958.
1995, 146, 261−296. (127) Hakansson, K.; Cooper, H. J.; Emmett, M. R.; Costello, C. E.;
(96) Jackson, G. S.; White, F. M.; Guan, S.; Marshall, A. G. J. Am. Soc. Marshall, A. G.; Nilsson, C. L. Anal. Chem. 2001, 73, 4530−4536.
Mass Spectrom. 1999, 10, 759−769. (128) McAlister, G. C.; Phanstiel, D.; Good, D. M.; Berggren, T.;
(97) Naito, Y.; Fujiwara, M.; Inoue, M. Int. J. Mass Spectrom. Ion Coon, J. J. Anal. Chem. 2007, 79, 3525−3534.
Processes 1992, 120, 179−192. (129) Mann, M.; Kelleher, N. L. Proc. Natl. Acad. Sci. 2008, 105,
(98) Vartanian, V. H.; Hadjarab, F.; Laude, D. A. Int. J. Mass 18132−18138.
Spectrom. 1995, 151, 175−187. (130) Xu, F.; Xu, Q.; Dong, X.; Guy, M.; Guner, H.; Hacker, T.; Ge, Y.
(99) Weisbrod, C. R.; Kaiser, N. K.; Skulason, G. E.; Bruce, J. E. Anal. Int. J. Mass Spectrom. 2011, 305, 95−102.
Chem. 2008, 80, 6545−6553. (131) Ryan, C. M.; Souda, P.; Bassilian, S.; Ujwal, R.; Zhang, J.;
(100) Brustkern, A. M.; Rempel, D. L.; Gross, M. L. J. Am. Soc. Mass Abramson, J.; Ping, P.; Durazo, A.; Bowie, J. U.; Hasan, S. S.; Baniulis,
Spectrom. 2008, 19, 1281−1285. D.; Cramer, W. A.; Faull, K. F.; Whitelegge, J. P. Mol. Cell. Proteomics
(101) Brustkern, A. M.; Rempel, D. L.; Gross, M. L. J. Am. Soc. Mass 2010, 791−803.
Spectrom. 2010, 21, 451−454. (132) Li, M.; Jiang, L.; Kelleher, N. L. J. Chromatogr., B 2009, 877,
(102) Kaiser, N. K.; Savory, J. J.; McKenna, A. M.; Quinn, J. P.; 3885−3892.
Hendrickson, C. L.; Marshall, A. G. Anal. Chem. 2011, 83, 6907−6910. (133) Pesavento, J. J.; Yang, H.; Kelleher, N. L.; Mizzen, C. A. Mol.
(103) Tolmachev, A. V.; Robinson, E. W.; Smith, R. D.; Pasa-Tolic, L. Cell. Biol. 2008, 28, 468−486.
J. Am. Soc. Mass Spectrom. 2011, 22, 1334−1342. (134) Ferguson, J. T.; Wenger, C. D.; Metcalf, W. W.; Kelleher, N. L.
(104) Tolmachev, A. V.; Robinson, E. W.; Wu, S.; Kang, H.; J. Am. Soc. Mass Spectrom. 2009, 20, 1743−1750.
Lourette, N. M.; Pasa-Tolic, L.; Smith, R. D. J. Am. Soc. Mass Spectrom. (135) Lee, J. E.; Kellie, J. F.; Tran, J. C.; Tipton, J. D.; Catherman,
2008, 19, 586−597. A. D.; Thomas, H. M.; Ahlf, D. R.; Durbin, K. R.; Vellaichamy, A.;
(105) Boldin, I. A.; Nikolaev, E. N. Rapid Commun. Mass Spectrom. Ntai, I.; Marshall, A. G.; Kelleher, N. L. J. Am. Soc. Mass Spectrom.
2010, 25, 122−126. 2009, 20, 2183−2191.
(106) Valeja, S. G.; Kaiser, N. K.; Xian, F.; Hendrickson, C. L.; (136) Pesavento, J. J.; Bullock, C. R.; LeDuc, R. D.; Mizzen, C. A.;
Rouse, J. C.; Marshall, A. G. Anal. Chem. 2011, 83, 8391−8395. Kelleher, N. L. J. Proteome Res. 2008, 283, 14927−14937.
(107) Misharin, A. S.; Zubarev, R. A.; Doroshenko, V. M. Rapid (137) Durbin, K. R.; Tran, J. C.; Zamdborg, L.; Sweet, S. M. M.;
Commun. Mass Spectrom. 2010, 24, 1931−1940. Catherman, A. D.; Lee, J. E.; Li, M.; Kellie, J. F.; Kelleher, N. L.
(108) Ledford, E. B.; Rempel, D. L.; Gross, M. L. Anal. Chem. 1984, Proteomics 2010, 10, 3589−3597.
56, 2744−2748. (138) Sampson, J. S.; Murray, K. K.; Muddiman, D. C. J. Am. Soc.
(109) Shi, S. D.-H.; Drader, J. J.; Freitas, M. A.; Hendrickson, C. L.; Mass Spectrom. 2009, 20, 667−673.
Marshall, A. G. Int. J. Mass Spectrom. 1999, 195/196, 591−598. (139) Lee, J. E.; Atkins, N.; Hatcher, N. G.; Zamdborg, L.; Gillette,
(110) Jefferies, J. B.; Barlow, S. E.; Dunn, G. H. Int. J. Mass Spectrom. M. U.; Sweedler, J. V.; Kelleher, N. L. Mol. Cell. Proteomics 2010, 9,
Ion Processes 1983, 34, 169−187. 285−297.
(111) Easterling, M. L.; Mize, T. H.; Amster, I. J. Anal. Chem. 1999, (140) Warder, S. E.; Tucker, L. A.; McLoughlin, S. M.; Strelitzer, T. J.;
71, 624−632. Meuth, J. L.; Zhang, Q.; Sheppard, G. S.; Richardson, P. L.;
(112) Muddiman, D. C.; Oberg, A. L. Anal. Chem. 2005, 77, 2406− Lesniewski, R.; Davidsen, S. K.; Bell, R. L.; Rogers, J. C.; Wang, J.
2414. J. Proteome Res. 2008, 7, 4807−4820.
(113) Rodgers, R. P.; White, F. M.; Hendrickson, C. L.; Marshall, (141) Armirotti, A. Curr. Anal. Chem. 2009, 5, 116−130.
A. G.; Andersen, K. V. Anal. Chem. 1998, 70, 4734−4750. (142) Han, X.; Aslanian, A.; Yates, J. R. III Curr. Opin. Chem. Biol.
(114) Gorshkov, M.; Good, D. M.; Lyutvinshiy, Y.; Yang, H.; 2008, 12, 483−490.
Zubarev, R. A. J. Am. Soc. Mass Spectrom. 2010, 21, 1846−1851. (143) Perkins, D. N.; Pappin, D. J. C.; Creasy, D. M.; Cottrell, J. S.
(115) Comisarow, M. B.; Marshall, A. G. Can. J. Chem. 1974, 52, Electrophoresis 1999, 20, 3551−3567.
1997−1999. (144) Eng, J. K.; McCormack, A. L.; Yates III, J. R. J. Am. Soc. Mass
(116) Beu, S. C.; Blakney, G. T.; Quinn, J. P.; Hendrickson, C. L.; Spectrom. 1994, 5, 976−989.
Marshall, A. G. Anal. Chem. 2004, 76, 5756−5761. (145) Brosch, M.; Swamy, S.; Hubbard, T.; Choudhary, J. Mol. Cell.
(117) Vining, B. A.; Bossio, R. E.; Marshall, A. G. Anal. Chem. 1999, Proteomics 2008, 962−970.
71, 460−467. (146) Sweet, S. M. M.; Jones, A. W.; Cunningham, D. L.; Heath,
(118) Xian, F.; Hendrickson, C. L.; Blakney, G. T.; Beu, S. C.; J. K.; Creese, A. J.; Cooper, H. J. J. Proteome Res. 2009, 8, 5475−5484.
Marshall, A. G. Anal. Chem. 2010, 82, 8807−8812. (147) Tolmachev, A. V.; Monroe, M. E.; Purvine, S. O.; Moore, R. J.;
(119) Qi, Y.; Thompson, C. J.; Van Ordan, S. L.; O’Connor, P. B. Jaitly, N.; Adkins, J. N.; Anderson, G. A.; Smith, R. D. Anal. Chem.
J. Am. Soc. Mass Spectrom. 2011, 22, 138−147. 2008, 80, 8514−8525.
(120) Lange, O.; Damoc, E.; Wieghaus, A.; Makarov, A. Proceedings of (148) Karpievitch, Y.; Stanley, J.; Taverner, T.; Huang, J.; Adkins,
the 59th ASMS Conference on Mass Spectrometry & Allied Topics, J. N.; Ansong, C.; Heffron, F.; Metz, T. O.; Qian, W.-J.; Yoon, H.;
Denver, CO, June 5−9, 2011. Smith, R. D.; Dabney, A. R. Bioinformatics 2009, 25, 2028−2034.
(121) Blakney, G. T.; Hendrickson, C. L.; Marshall, A. G. Int. J. Mass (149) Bereman, M. S.; Egertson, J. D.; MacCoss, M. J. Proteomics
Spectrom. 2011, 306, 246−252. 2011, 11, 2931−2935.

717 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719


Analytical Chemistry Review

(150) Porteus, B.; Kocharunchitt, C.; Nilsson, R. E.; Ross, T.; (179) Zou, Y.; Tiller, P.; Chen, I.-W.; Beverly, M.; Hochman, J. Rapid
Bowman, J. P. Appl. Microbiol. Biotechnol. 2011, 90, 407−416. Commun. Mass Spectrom. 2008, 22, 1871−1881.
(151) Plazas-Mayorca, M. D.; Zee, B. M.; Young, N. L.; Fingerman, (180) Bahr, U.; Aygun, H.; Karas, M. Anal. Chem. 2009, 81, 3173−
I. M.; LeRoy, G.; Briggs, S. D.; Garcia, B. A. J. Proteome Res. 2009, 8, 3179.
5367−5374. (181) Taoka, M.; Yamauchi, Y.; Nobe, Y.; Masaki, S.; Nakayama, H.;
(152) Sweet, S. M. M.; Li, M.; Thomas, P. M.; Durbin, K. R.; Ishikawa, H.; Takahashi, N.; Isobe, T. Nucleic Acids Res. 2009, 37,
Kelleher, N. L. J. Biol. Chem. 2010, 285, 32778−32786. 1−14.
(153) Bumpus, S. B.; Evans, B. S.; Thomas, P. M.; Ntai, I.; Kelleher, (182) Turner, K. B.; Brinson, R. G.; Yi-Brunozzi, H. Y.; Rausch, J. W.;
N. L. Nat. Biotechnol. 2009, 27, 951−955. Miller, J. T.; Le Grice, S. F. J.; Marino, J. P.; Fabris, D. Nucleic Acids
(154) Boyne, M. T. II; Garcia, B. A.; Li, M.; Zamdborg, L.; Wenger, Res. 2008, 36, 2799−2810.
C. D.; Babal, S.; Kelleher, N. L. J. Proteome Res. 2009, 8, 374−379. (183) Turner, K. B.; Young, Y.-B.; Hye.; Brinson, R. G.; Marino, J. P.;
(155) Garcia, B. A.; Thomas, C. E.; Kelleher, N. L.; Mizzen, C. A. Fabris, D.; Le Grice, S. F. J. RNA 2009, 15, 1605−1613.
J. Proteome Res. 2008, 7, 4225−4236. (184) Turner, K. B.; Kohlway, A. S.; Hagan, N. A.; Fabris, D.
(156) Ong, S.-E.; Blagoev, B.; Kratchmarova, I.; Kristensen, D. B.; Biopolymers 2008, 91, 283−295.
Steen, H.; Pandey, A.; Mann, M. Mol. Cell. Proteomics 2002, 1, 376− (185) Yu, E. T.; Hawkins, A.; Eaton, J.; Fabris, D. Proc. Natl. Acad.
586.
Sci. 2008, 105, 12248−12253.
(157) Manes, N. P.; Estep, R. D.; Mottaz, H. M.; Moore, R. J.;
(186) Weidner, S. M.; Trimpin, S. Anal. Chem. 2010, 82, 4811−4829.
Clauss, T. R.; Monroe, M. E.; Du, X.; Adkins, J. N.; Wong, S. W.;
(187) Pernot, P.; Carrasco, N.; Thissen, R.; Schmitz-Afonso, I. Anal.
Smith, R. D. J. Proteome Res. 2008, 7, 960−968.
Chem. 2010, 82, 1371−1380.
(158) Wu, J.; Warren, P.; Shakey, Q.; Sousa, E.; Hill, A.; Ryan, T.;
(188) Wienhofer, I. C.; Luftmann, H.; Studer, A. Macromolecules
He, T. Proteomics 2010, 10, 2224−2234.
(159) Dayon, L.; Pasquarello, C.; Hoogland, C.; Sanchez, J.-C.; 2011, 44, 2510−2523.
Scherl, A. J. Proteomics 2010, 73, 769−777. (189) Kaczorowska, M.; Cooper, H. J. J. Am. Soc. Mass Spectrom.
(160) Zhang, Y.; Ficarro, S. B.; Li, S.; Marto, J. A. J. Am. Soc. Mass 2008, 19, 1312−1319.
Spectrom. 2009, 20, 1425−1434. (190) Kaczorowska, M.; Cooper, H. J. J. Am. Soc. Mass Spectrom.
(161) Zybailov, B.; Coleman, M. K.; Florens, L.; Washburn, M. P. 2009, 20, 2238−2247.
Anal. Chem. 2005, 77, 6218−6224. (191) Kaczorowska, M.; Cooper, H. J. J. Am. Soc. Mass Spectrom.
(162) Taylor, N. S.; Weber, R. J. M.; Southam, A. D.; Payne, T. G.; 2009, 20, 674−681.
Hrydziuszko, O.; Arvanitis, T. N.; Viant, M. R. Metabolomics 2009, 5, (192) Miladinovic, S. M.; Kaeser, C. J.; Knust, M. M.; Wilkins, C. L.
44−58. Int. J. Mass Spectrom. 2011, 301, 184−194.
(163) Denkert, C.; Budczies, J.; Weichert, W.; Wohlgemuth, G.; (193) Nasioudis, A.; Heeren, R. M. A.; van Doormalen, I.; de Wijs-
Scholz, M.; Kind, T.; Niesporek, S.; Noske, A.; Buckendahl, A.; Dietel, Rot, N.; Van den Brink, O. F. J. Am. Soc. Mass Spectrom. 2011, 22,
M.; Fiehn, O. Mol. Cancer 2008, 7, 72. 837−844.
(164) Takahashi, H.; Kai, K.; Shinbo, Y.; Tanaka, K.; Ohta, D.; (194) Song, J.; van Velde, J. W.; Vertommen, L. L. T.; van der Ven,
Oshima, T.; Altaf-UI-Amin, M.; Kurokawa, K.; Ogasawara, N.; Kanaya, S. L. G. J.; Heeren, R. M. A.; Van den Brink, O. F. Macromolecules 2010,
Anal. Bioanal. Chem. 2008, 391, 2769−2782. 43, 7082−7089.
(165) Baidoo, E. E. K.; Benke, P. I.; Neususs, C.; Pelzing, M.; (195) Marshall, A. G.; Rodgers, R. P. Proc. Natl. Acad. Sci. U.S.A.
Kruppa, G.; Leary, J. A.; Keasling, J. D. Anal. Chem. 2008, 80, 3112− 2008, 105, 18090−18095.
3122. (196) Rodgers, R. P.; McKenna, A. M. Anal. Chem. 2011, 83, 4665−
(166) Dunn, W. B.; Broadhurst, D.; Brown, M.; Baker, P. N.; 4687.
Redman, C. W. G.; Kenny, L. C.; Kell, D. B. J. Chromatogr., B 2008, (197) Fernandez-Lima, F. A.; Becker, C.; McKenna, A. M.; Rodgers,
871, 288−298. R. P.; Marshall, A. G.; Russell, D. H. Anal. Chem. 2009, 81, 9941−
(167) Kamleh, M. A.; Hobani, Y.; Dow, J. A. T.; Watson, D. G. FEBS 9947.
Lett. 2008, 582, 2916−2922. (198) Betancourt, S. S.; Ventura, G. T.; Pomerantz, A. E.; Viloria, O.;
(168) Werner, E.; Croixmarie, V.; Umbdenstock, T.; Ezan, E.; Dubost, F. X.; Zuo, J.; Monson, G.; Bustamante, D.; Purcell, J. M.;
Chaminade, P.; Tabet, J.-C.; Junot, C. Anal. Chem. 2008, 80, 4918− Nelson, R. K.; Rodgers, R. P.; Reddy, C. M.; Marshall, A. G.; Mullins,
4932. O. C. Energy Fuels 2009, 23, 1178−1188.
(169) Jin, Z.; Daiya, S.; Kenttamaa, H. I. Int. J. Mass Spectrom. 2011, (199) McKenna, A. M.; Purcell, J. M.; Rodgers, R. P.; Marshall, A. G.
301, 234−239. Energy Fuels 2009, 23, 2122−2128.
(170) Murphy, R. C.; Gaskell, S. J. J. Biol. Chem. 2011, 286, 25427− (200) Pinkston, D. S.; Duan, P.; Gallardo, V. A.; Habicht, S. C.; Tan,
25433.
X.; Qian, K.; Gray, M.; Mullen, K.; Kenttamaa, H. I. Energy Fuels 2009,
(171) Huan, H.; Emmett, M. R.; Nilsson, C. L.; Conrad, C. A.;
23, 5564−5570.
Marshall, A. G. Int. J. Mass Spectrom. 2010, 305, 116−119.
(201) Purcell, J. M.; Merdrignac, I.; Rodgers, R. P.; Marshall, A. G.;
(172) Huan, H.; Nilsson, C. L.; Emmett, M. R.; Ji, Y.; Marshall, A. G.;
Kroes, R. A.; Moskai, J. R.; Colman, H.; Lang, F. F.; Conrad, C. A. Gauthier, T.; Guibard, I. Energy Fuels 2010, 24, 2257−2265.
Glycoconjugate J. 2010, 27, 27−38. (202) Smith, D. F.; Rodgers, R. P.; Rahimi, P.; Teclemariam, A.;
(173) Huan, H.; Rodger, R. P.; Marshall, A. G.; Hsu, C. S. Energy Marshall, A. G. Energy Fuels 2009, 23, 314−319.
Fuels 2011, 26, 4770−4775. (203) Mullins, O. C.; Sheu, E. Y.; Hammami, A.; Marshall, A. G., Eds.
(174) Lane, A. N.; Fan, T. W.-M.; Xie, Z.; Moseley, H. N. B.; Higashi, Asphaltenes, Heavy Oils, and Petroleomics; Springer: New York, 2007.
R. M. Anal. Chim. Acta 2009, 651, 201−208. (204) McKenna, A. M.; Blakney, G. T.; Xian, F.; Glaser, P. B.;
(175) Garnier, N.; Rolando, C.; Hotje, J. M.; Tokarski, C. Int. J. Mass Rodgers, R. P.; Marshall, A. G. Energy Fuels 2010, 24, 2939−2946.
Spectrom. 2009, 284, 47−56. (205) McKenna, A. M.; Purcell, J. M.; Rodgers, R. P.; Marshall, A. G.
(176) Komaniecka, I.; Choma, A.; Lindner, B.; Holst, O. Chem. Energy Fuels 2010, 24, 2929−2938.
Eur. J. 2009, 16, 2922−2929. (206) Hughey, C. A.; Minardi, C. S.; Galasso-Roth, S. A.; Paspalof,
(177) Pol, J.; Vidova, V.; Hyotylainen, T.; Volny, M.; Novak, P.; G. B.; Mapolelo, M. M.; Rodgers, R. P.; Marshall, A. G.; Ruderman,
Strohalm, M.; Kostiainen, R.; Havlicek, V.; Wiedmer, S.; Holopainen, D. L. Rapid Commun. Mass Spectrom. 2008, 22, 3968−3976.
J. M. Plos ONE 2011, 6, 1−9. (207) Mapolelo, M. M.; Rodgers, R. P.; Blakney, G. T.; Yen, A. T.;
(178) Vidova, V.; Pol, J.; Volny, M.; Novak, P.; Havlicek, V.; Asomaning, S.; Marshall, A. G. Int. J. Mass Spectrom. 2011, 300, 149−
Wiedmer, S.; Holopainen, J. M. J. Lipid Res. 2010, 51, 2295−2299. 157.

718 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719


Analytical Chemistry Review

(208) Mapolelo, M. M.; Stanford, L. A.; Rodgers, R. P.; Yen, A. T.;


Debord, J. D.; Asomaning, S.; Marshall, A. G. Energy Fuels 2009, 23,
349−355.
(209) Headley, J. V.; Barrow, M. P.; Peru, K. M.; Derrick, P. J.
J. Environ. Sci. Health, Part A 2011, 46, 844−854.
(210) Headley, J. V.; Peru, K. M.; Janfada, A.; Fahlman, B.; Gu, C.;
Hassan, S. Rapid Commun. Mass Spectrom. 2011, 25, 459−462.
(211) Headley, J. V.; Peru, K. M.; Mishra, S.; Meda, V.; Dalai, A. K.;
McMartin, D. W.; Mapolelo, M. M.; Rodgers, R. P.; Marshall, A. G.
Rapid Commun. Mass Spectrom. 2010, 24, 3121−3126.
(212) Juyal, P.; McKenna, A. M.; Yen, A. T.; Rodgers, R. P.; Reddy,
C. M.; Nelson, R. K.; Andrews, A. B.; Atolia, E.; Allenson, S. J.;
Mullins, O. C.; Marshall, A. G. Energy Fuels 2011, 25, 172−182.
(213) Pomerantz, A. E.; Ventura, G. T.; McKenna, A. M.; Canas,
J. A.; Auman, J.; Koerner, K.; Curry, D.; Nelson, R. K.; Reddy, C. M.;
Rodgers, R. P.; Marshall, A. G.; Peters, K. E.; Mullins, O. C. Org.
Geochem. 2010, 41, 812−821.
(214) Kellmann, M.; Muenster, H.; Zomer, P.; Mol, H. J. Am. Soc.
Mass Spectrom. 2009, 20, 1464−1476.
(215) Krauss, M.; Singer, H.; Hollender, J. Anal. Bioanal. Chem. 2010,
397, 943−951.
(216) Helbling, D. E.; Hollender, J.; Kohler, H.-P. E.; Singer, H.;
Fenner, K. Environ. Sci. Technol. 2010, 44, 6621−6627.
(217) Liou, J. S.-C.; Szostek, B.; DeRito, C. M.; Madsen, E. L.
Chemosphere 2010, 80, 176−183.
(218) van Leerdam, J. A.; Hogenboom, A. C.; van der Kooi, M. M. E.;
de Voogt, P. Int. J. Mass Spectrom. 2009, 282, 99−107.
(219) Nam, S.; Joo, S.; Kim, S.; Baek, N.-I.; Choi, H.-K.; Park, S.
J. Plant Biol. 2008, 51, 373−378.
(220) Schmidt, F.; Elvert, M.; Koch, B. P.; Witt, M.; Hinrichs, K.-U.
Geochim. Cosmochim. Acta 2009, 73, 3337−3358.
(221) Gonsior, M.; Zwartjes, M.; Cooper, W. J.; Song, W.; Ishida,
K. P.; Tseng, L. Y.; Jeung, M. K.; Rosso, D.; Hertkorn, N.; Schmitt-
Kopplin, P. Water Res. 2011, 45, 2943−2953.
(222) Bhatia, M. P.; Das, S. B.; Longnecker, K.; Charette, M. A.;
Kujawinski, E. B. Geochim. Cosmochim. Acta 2010, 74, 3768−3784.
(223) Giardina, M.; Siek, K.; Kowalski, J. In Proceedings of the 58th
ASMS Conference on Mass Spectrometry & Allied Topics, Salt Lake City,
Utah, May 23−27, 2010.

719 dx.doi.org/10.1021/ac203191t | Anal. Chem. 2012, 84, 708−719

You might also like