Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Review

pubs.acs.org/ac

NMR Spectroscopy for Metabolomics and Metabolic Profiling


Cynthia K. Larive,*,† Gregory A. Barding, Jr.,‡ and Meredith M. Dinges†

Department of Chemistry, University of California−Riverside, Riverside, California 92521, United States


Chemistry and Biochemistry Department, California State Polytechnic University, Pomona, California 91768, United States
CONTENTS
Applications of NMR Metabolomics 133
Biofluids 134
Downloaded via INST NAC DE TECNOLOGIA INDUSTRIAL on September 23, 2022 at 11:56:07 (UTC).

Cells and Tissues 134


See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Plants 134
Natural Products 134
Environmental Interactions 135
Food and Beverages 135
Experimental Design 135
Sample Storage and Preparation 136
Extraction 136
Development of New Methods and Experiments: 1H
NMR 136
Relaxation Effects 136
New Pulse Sequences 137
Fast and Ultrafast NMR Experiments 137
Probe Developments 137
High-Resolution Magic Angle Spinning (HR-MAS) 138
Development of New Methods and Experiments: Figure 1. Growth in publications on NMR metabolomics/
Nuclei Other than 1H 138 metabonomics since 2000. These results were obtained from a topic
Isotope Labeling 138 search of all documents on the Web of Knowledge using the keywords
Metabolic Flux Analysis 139 “NMR and metabonomic*” and “NMR and metabolomics*”.
Hyperpolarization and DNP 140
Data Processing and Chemometrics 140 illustrates the rapid growth in publications on topics that
Spectral Processing 141 include the keywords NMR and metabolomics/metabonomics
Resonance Assignments and Databases 142 since the year 2000. Due to space limitations, this Review
Quantitation 142 covers only papers published between 2011 and the first half
Chemometrics 142 of 2014.
Perspective
Author Information
Corresponding Author
143
144
144
■ APPLICATIONS OF NMR METABOLOMICS
The omics revolution in systems biology has provided an
Notes 144
unprecedented level of understanding of how organisms
Biographies 144
respond to environmental factors such as diet, aging, and
Acknowledgments 144
disease. As the end products of transcription and translation,
References 144
small molecule metabolites can be considered as the output
of the biological system, in essence a molecular phenotype.

A pplications of NMR for metabolomics and metabolic


profiling continue to grow rapidly as does the refinement
of methods for the measurement, analysis, and interpretation of
Because metabolites can impact gene transcription through
feedback loops, the flow of information from the genome
through the transcriptome and proteome to the metabolome is
complex data sets. Metabolomics (metabonomics) is a set of bidirectional (Figure 2).1,2 Though a comprehensive review
global measurements performed on biological samples with of NMR metabolomics applications is beyond the scope of
the goal of quantifying as many metabolites as possible and this Review, the impact of this approach can be appreciated by
evaluating changes in metabolite levels as a result of an applied a survey of recently published review articles. The section
stress. Metabolic profiling experiments follow a more limited below provides an overview of recent reviews describing appli-
set of metabolites often through specific pathways. NMR is also cations of NMR-based metabolomics. In addition, the review by
well suited for metabolite fingerprinting, which involves the
comprehensive and simultaneous analysis of a wide variety of Special Issue: Fundamental and Applied Reviews in Analytical
compounds. For the purpose of this Review, we do not distin- Chemistry 2015
guish between metabolomics and metabonomics and have
elected to use the term metabolomics throughout. Figure 1 Published: November 6, 2014

© 2014 American Chemical Society 133 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

Figure 2. Complex interactions of functional levels (genome, transcriptome, proteome, and metabolome) in biological systems. Bidirectional flows of
biological information are observed between the genome, transcriptome, proteome, and metabolome. The complex interaction of components from
all the functional levels and the environment produces the phenotype, the output of the system measured in systems-level metabolomics and systems
biology. Adapted from ref 1 with permission of The Royal Society of Chemistry, copyright 2011.

Bothwell and Griffin provides an excellent introduction to mixtures, was also demonstrated and validated as a rapid and
biological NMR.3 nondestructive tool for forensic analysis.12
Biofluids. Biofluids are an attractive sample matrix for Cells and Tissues. In addition to the global metabolic view
metabolomics studies because they can be obtained in a provided by blood and urine, sampling of intact tissues can
noninvasive (e.g., saliva, urine) or minimally invasive manner provide a more specific, local response to disease. For example
(e.g., blood plasma or serum, cerebrospinal fluid). NMR blood serum, bronchoalveolar lavage fluid, and excised lung
metabolomics studies using biofluids offer potential for a tissue were used to identify metabolic biomarkers of sepsis in
deeper understanding of disease pathogenesis and the rats.13 Another recent review documents the progress made in
identification of metabolic biomarkers useful for disease the application of NMR and magnetic resonance spectroscopic
diagnosis or treatment monitoring.4−7 Because of the aberrant imaging (MRSI) to detection, diagnosis, and characterization of
metabolism of cancer cells involving enhanced glucose uptake human prostate cancer.14 NMR metabolomics was used to
and glycolytic activity, a shift in the TCA cycle from oxidation investigate the effects of environmental stressors associated
to lipogenesis, increased glutaminolysis, and nucleotide biosyn- with biofilm development by Staphlococcus epidermis, strongly
thesis, metabolic screening of biofluids has the potential to implicating a central role for the TCA cycle.15 NMR studies on
contribute to early cancer screening, improved diagnostic the effect, mode of action, delivery, and toxicity of therapeutic
accuracy, and prediction of a patient’s response to treatment.8 drugs has been recently reviewed.16 NMR metabolomics also
For example, urine metabolic profiling shows excellent promise has potential to provide a thorough assessment of the toxicity
as a screening tool for bladder cancer.9 and mechanisms of action of new nanomaterial-based
For a metabolic biomarker to be clinically useful, its level therapeutics.17
must clearly associate with disease risk or progression, should Plants. Plant metabolomics studies have been used to
be insensitive to variables like ethnicity, diet, and location, and address the effects of genotype, ecotype, and environmental
should not vary too much over the short-term within an stressors such as drought and submergence18−20 and can
individual. To assess the sources of variation among individuals, provide a metabolic phenotype in chemical genomics studies in
blood plasma and urine 1H NMR spectra were measured for plants.21 The application of 1H NMR spectroscopy to plant
samples obtained from 154 healthy Caucasian, postmenopausal metabolomics is in many ways more complicated than biofluid
female twins (identical and nonidentical).10 Thirty four of the analysis and requires consideration of whether the whole plant,
identical twins provided two samples over the space of several a portion of the plant (i.e., leaves, roots, or fruit), or a single cell
months in this study. On the basis of these results, Nicholson type is most relevant to the question being explored.22 Most
et al. estimate that stable variation derived from familial and plant metabolomics studies are carried out on plant tissue
individual-environmental factors accounts for on average 60% extracts;18,22 however, the review by Serra et al. discusses the
of the biological variation in the 1H NMR spectra of plasma and use of solid state NMR for the characterization of the insoluble
47% of the variation in the urine spectra. The authors postulate protective biopolymers cutin and suberin.23 In addition to their
that clinically predictive metabolic variation potentially useful primary metabolome, plants also produce an immense number
for biomarker discovery lies within this stable component. In of secondary metabolites (∼200 000) that allow them to
an effort to catalog the metabolic components of urine, a interact with beneficial or harmful organisms. Leiss et al.
comprehensive study of the human urine metabolome using provide an overview on the use of NMR metabolomics to
NMR, GC/MS, ICPMS, and direct infusion- and LC-MS/MS identify compounds important for host plant resistance to
reported the identification of 445 and quantification of 378 western flower thrips (Frankliniella occidentalis).24
unique urine metabolites or metabolic species, with 179 Natural Products. NMR metabolomics approaches are also
metabolites detected using NMR.11 The potential of 1H finding use in discovery-oriented natural products chemistry.25
NMR metabolic profiling to identify trace quantities of body Comparison of high-resolution 2D NMR spectra of unfrac-
fluids (blood, urine, saliva, and semen,) singly or in binary tionated sample extracts can facilitate the detection and
134 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

identification of novel compounds that might be lost during


chromatographic fractionation because of their low abundance,
chemical properties, or instability. Activity guided fractionation,
a common approach in natural products research, also fails
when several biogenic small molecules (BSMs) act synergisti-
cally. Through the statistical analysis of NMR spectra of
complex mixtures of BSMs, unique spectral features can be
identified and correlated to a phenotype or biological property
of interest.26
Environmental Interactions. Environmental metabolo-
mics addresses the effects of complex environmental stressors
including abiotic factors (e.g., exposure to xenobiotic com-
pounds and temperature shifts) and biotic factors (e.g.,
herbivory and competition) and can be used to elucidate
their biochemical modes of action.27 As NMR metabolomics
studies using samples derived from human patients can be a
powerful tool for the discovery of disease biomarkers, metabolic
shifts in terrestrial or aquatic organisms are potential early
bioindicators of environmental stress. To integrate the results
of genomic, transcriptomic, and metabolomics data, Ogata et al.
introduced ECOMICS, a public domain web-based toolkit that
allows for monitoring of biomass metabolism and facilitates
understanding of relationships between molecular and micro-
bial elements.28
Food and Beverages. Though applications of NMR
metabolomics are most prevalent in studies using biofluids or
samples derived from living organisms, this approach has also
been used to address a diverse range of applications involving
other types of complex samples. For example, NMR
metabolomics has been used to assess the quality of plant
herbal products and juices.29 For wines, NMR metabolomics
measurements can reflect the grape variety and age of the
vintage, as well as provide insights to the “terroir”, which can
encompass the effects of geographic region, climate, soil,
agricultural practices, and fermentation organisms, parameters
well-known to impact the taste of wine as well as its metabolic
profile.30 The application of NMR metabolomics using wine,
juice, and milk samples has been described for student
instruction in a variety of courses while also illustrating how
students can use 1H NMR metabolic profiling to follow the
utilization of glucose by yeast.31 Figure 3. Work flow for clinical metabolomics/metabolic NMR

■ EXPERIMENTAL DESIGN
A typical metabolomics workflow is summarized diagrammati-
spectroscopy. Samples are collected in a uniform way to minimize
variability and are analyzed by an NMR profile to collect data on all
metabolites potentially present in the sample. Pattern recognition
approaches (PRA) include principal component analysis, partial least-
cally in Figure 3.32 The study begins with consideration of squares discriminant analysis, orthogonal projections to latent
factors such as determining the number of subjects, establishing structures, heat map, support vector machines, the random forests
appropriate controls, the types of samples that will be provided, method, and other modes aiming to highlight underlying trends and
how frequently samples will be taken, and how the samples will visualization tools such as contribution. Trend and box plots are used
be stored. The collected samples undergo preparation for NMR to further evaluate these. Receiver operating characteristic (ROC)
analysis, and the NMR spectra are recorded according to a curves are generally considered the method of choice for evaluating the
standardized protocol selected by the investigator. The NMR performance of potential biomarkers. The markers are eventually
data sets are then subjected to statistical analyses to identify placed in a metabolic pathway to provide insight on the biochemical
variance between the test groups and those NMR resonances phenomena. Reprinted from ref 32 with permission from John Wiley
& Sons Inc., copyright 2013.
important for distinguishing the sample groupings. Once key
metabolites are identified, a univariate analysis can be
performed to establish trends in metabolite levels as a function Athersuch have reported typical experimental protocols for
of a perturbation (i.e., dose, time) or to quantify differences NMR metabolomics analyses of biofluids, tissues, cell extracts,
between control and treatment samples. After significance has and culture media.33 A comparison study of technical replicates
been established, metabolic pathways are examined to under- of human urine samples at two universities using 5 mm and
stand the biochemical basis for the effect. 3 mm probes revealed the importance of parameter stand-
Proper experimental design is critical to obtaining meta- ardization in producing reliable and reproducible quantitative
bolomics data that addresses the experimental hypothesis and NMR metabolomics data.34 An important finding of this study
can be translated to other laboratories or situations. Keun and is that previously unrecognized parameters, especially those
135 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

related to solvent suppression, can have a dramatic effect on the


results. Though the metabolomics community has not yet
arrived at a set of standardized methods, standardization of
controlled vocabularies (or ontologies) and agreement on
reporting standards for the experimental methods and bio-
logical data facilitate data exchange.1 An important step toward
reducing interlaboratory variability, verifying method accuracy,
and validation of new measurement methods is the availability
of a standard reference material SRM 1950 “Metabolites
in Human Plasma” by the National Institute of Standards
and Technology (NIST).35 SRM 1950 is provided with
comprehensive metabolomics characterization using GC/MS,
LC-MS/MS, and NMR.
Sample Storage and Preparation. As in almost any
analysis, reproducible and effective methods of sample storage,
preservation, and preparation are critical to the outcome of
NMR metabolomics studies. Factors affecting the inter- and
intrasubject variability in human urine samples, including
storage conditions and diet, were investigated.36 Bacterial
metabolites were minimized by storing the samples under
refrigeration, and diet standardization attenuated the effects of
diet culture and cohabitation, though gender-specific differ-
ences remained. The effects of sample handling and storage
conditions on human plasma samples for NMR metabolomics
studies have also been investigated.37 Due to the high protein
content of blood plasma or serum samples, which obscures
the resonances of small molecule metabolites, it is typically
necessary to physically remove the proteins by precipitation or
ultrafiltration or to attenuate the intensity of protein resonances
in the 1H NMR spectrum based on their faster rates of
T2 relaxation. Gowda and Raftery compared these approaches
and found that precipitation of blood serum proteins using
methanol gave the best performance, producing significantly
higher levels of some metabolites than other approaches.38 An
optimized method for attenuating intersample differences in the
pH and dication concentrations in NMR spectra of rat, mouse,
and human urine samples has also been reported.39
Extraction. Extraction is a critical component of sample
preparation protocols for cell and tissue analysis,32 but it
can also be slow and labor-intensive. Matheus et al. report a Figure 4. 1H NMR spectra of the “wash”, “elute 1”, and “elute 2”
streamlined approach using sonication to disrupt cell mem- fractions compared with the reference (nonfractionated) profile from
branes or tissue structures during the extraction step.40 A study the same urine sample. The blue profile was reconstructed by adding
exploring extraction protocols using solvents with varying the “wash”, “elute 1”, and “elute 2” subprofiles. The aromatic and
polarities for NMR metabolomics of adherent mammalian aliphatic regions are shown in (a) and (b), respectively. Reprinted
cells demonstrated that a higher proportion of hydrophilic from ref 42 with kind permission from Springer Science and Business
metabolites was obtained with a solvent of methanol/ Media, copyright 2012.
chloroform/water.41 Compared with liquid−liquid extraction
methods, solid phase extraction (SPE) can be useful for strategy for structure elucidation and compound identification
in metabolomics studies.43−45


fractionating complex samples prior to measurement of NMR
spectra and can be more amenable to automation. SPE-NMR
was used to isolate 3 fractions from urine samples based on DEVELOPMENT OF NEW METHODS AND
differences in polarity and improved the selectively of the NMR EXPERIMENTS: 1H NMR
measurements. Figure 4 shows the 1H NMR spectra obtained Because of its high sensitivity and natural abundance and its
for urine samples applied to hydrophilic−lipophilic balance nearly ubiquitous presence in organic metabolites, many
(HLB) SPE cartridges and collected into three fractions which metabolomics studies rely on the measurement of 1H NMR
were analyzed separately: wash (load + 3 wash volumes of spectra. In this section, efforts to improve the reproducibility,
0.5 mL of water), elute 1 (3 elutions with 0.5 mL of 10% throughput, sensitivity, and selectivity of 1H NMR meta-
methanol/90% water), and elute 2 (3 elutions of 0.5 mL of bolomics measurements are summarized.
methanol).42 Mass-guided SPE-trapping of selected compounds Relaxation Effects. Most NMR metabolomics experiments,
for NMR measurements were used in conjunction with HPLC- and especially those performed on biofluids, depend on effec-
FTMS to elucidate the structures of 36 phenolic conjugates in tive suppression of the water resonance. One of the most pop-
human urine following a single bolus intake of black or green ular methods of solvent suppression is the 1D-NOESY pulse
tea.43 Hyphenation of LC-MS-SPE-NMR has proven a useful sequence, because of its robustness and ease of implementation.
136 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

Figure 5. Schematic diagram for MaxEnt. MaxEnt reconstruction begins with empirical data and a preliminary trial spectrum f (typically a blank
spectrum). Spectrum f is inverted (DFT−1) to create “mock” data (m) that is compared with the empirical data (d). An update to the trial spectrum
is computed by searching along the gradients of the entropy and the constraint (the agreement between the empirical and mock data).
The algorithm converges to the unique MaxEnt solution when the gradient of the objective function Q = S − λC is zero and the gradients of S and C
are antiparallel. Reprinted from ref 52. Copyright 2014 American Chemical Society.

Parameter optimization and the mechanism of 1D NOESY dimension is limited. One way to overcome this limitation is
for water suppression, which in part depends on differences in through fast NMR experiments which use nonuniform sampling
the T1 relaxation properties of water and the metabolites of with maximum entropy (MaxEnt) reconstruction, a non-Fourier
interest, have been recently reviewed.46 In a different app- method of spectrum analysis, illustrated in Figure 5.52 Rai and
lication of the NOESY experiment, Farooq et al. report the use Sinha report the use of nonlinear sampling, forward maximum
of supercooled water in 1.4 mm capillaries to measure high entropy reconstruction, and the J-compensated heteronuclear
quality 2D NOESY spectra of small molecule metabolites single quantum coherence (HSQC) pulse sequence to achieve a
at −15 °C.47 At this temperature, the solvent viscosity is 3−4 22-fold reduction in the data collection time for body fluid
times higher than water at room temperature affecting the rate samples without any compromise in the quantitation of low
of proton cross relaxation and producing strong NOESY abundance metabolites.53
correlations that can be used in concert with 2D experiments Though maximum entropy reduces the time required for
based on J-coupling for resonance assignment and metabolite multidimensional NMR experiments by reducing the number
identification. of data points acquired, ultrafast NMR experiments which
New Pulse Sequences. To overcome problems of res- grew out of magnetic resonance imaging techniques, allow
onance overlap in the 1H NMR spectra of complicated samples, the acquisition of 2D NMR spectra in a single scan.54 These
Martin-Pastor described a one-dimensional singlet-filtered ultrafast 2D NMR experiments open the door to high
experiment that edits complex spectra to reveal only the resolution and high throughput 2D NMR-based metabolomics
peaks of singlet resonances and weakly coupled signals.48 studies.55 An ultrafast heteronuclear 2D J-resolved spectros-
Experimental schemes that produce high quality 2D J-resolved copy pulse sequence has been reported.56 As an extension of
spectroscopy and spin−echo correlated spectroscopy (SECSY) this experiment, an ultrafast 3D UFJCOSY pulse sequence was
spectra in inhomogeneous magnetic fields have also been described that produces COSY-type correlations with hetero-
reported and demonstrated to be useful for in vivo measure- nuclear couplings displayed in the third dimension.57 This
ments.49,50 Longitudinal relaxation enhancements using experiment allows direct measurement of the level of isotopic
selective excitation and refocusing pulses were demonstrated enrichments, which are critical for metabolic flux experiments.58
to increase the sensitivity of in vivo detection of mouse brain In addition to greatly reducing the acquisition time, these
metabolites by magnetic resonance spectroscopy (MRS).51 ultrafast experiments are also widely applicable for quantita-
Fast and Ultrafast NMR Experiments. Pulsed NMR tive NMR measurements.59 Coupled with stable isotope
experiments typically rely on the discrete Fourier transform labeling and polarization methods that enhance sensitivity,
(DFT) to convert digitized time domain free induction decays this breakthrough promises to revolutionize in vivo NMR
(FIDs) to frequency domain data. Multidimensional NMR spectroscopy.
spectra using DFT require uniform sampling intervals with Probe Developments. The NMR probe contains the
sufficiently small time increments in the indirect dimension hardware elements necessary to apply radio frequency pulses
to satisfy the Nyquist condition and long evolution times for to the sample and to detect the resultant signal prior to
high resolution. As a result, multidimensional NMR spectra often amplification and digitization. Therefore, probe design and
require long acquisition times and resolution in the indirect optimization is an avenue frequently explored to improve
137 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

detection sensitivity or to enable new types of measurements. than 1 Hz.69 This technology was used to obtain 1H NMR
Renslow et al. describe a biofilm microreactor seated on a spectra at 18.8 and 23.5 T for two strains of C. elegans
custom built NMR probe that allows for simultaneous nematodes highlighting the potential of NMR microprobe
electrochemical and NMR measurements at the microscale.60 technologies for metabolic screening of small model organ-
Other advances in probe design include development of a isms.70 To minimize heating from eddy currents and reduce the
13
C-optimized probe with a 35 μL sample volume constructed centrifugal force felt by the worms, a spinning frequency of
from a high temperature superconducting oxide deposited on a 350 Hz was used.


sapphire substrate.61 To facilitate experiments involving hetero-
nuclear hyperpolarization at low fields, a single input, double- DEVELOPMENT OF NEW METHODS AND
tuned probe circuit has been developed that enables double EXPERIMENTS: NUCLEI OTHER THAN 1H
resonance NMR to be performed with single channel consoles
using PANORAMIC (Precession And Nutation for Observing A fundamental advantage of using NMR to probe biological
Rotations At Multiple Intervals about the Carrier) waveforms.62 systems is the ability to measure a wide variety of spin 1/2
Compared with traditional 5 mm NMR probes, microcoil nuclei, including (but not limited to) the biologically relevant
1
probes offer improved mass sensitivity. The application of H, 13C, 31P, and 15N in addition to the pharmaceutically
microcoil NMR for metabolomics of urine and serum samples, relevant 19F. Of these, 1H, 19F, and 31P are found at high
including the effect of concentrating biofluid samples on percentages of natural abundance. The lower natural abundance
metabolite quantification, has been investigated using a probe of 13C and 15N can be useful in measuring enrichment due to
designed for flow injection analysis.63 A stripline NMR probe catabolism of labeled precursors as a function of specific bio-
has been integrated into a microfluidic flow system allowing logical processes or pathways. More specifically, NMR can
electrochemical generation of reactive products concentrated easily be used to monitor the positional enrichment of a
via SPE before measurement of the NMR spectrum in the molecule for both 13C and 15N NMR to establish which
150 nL active volume.64 Though microfluidic chip-based flow pathways are being activated or inactivated. The sensitivity
probes are potentially attractive for high-throughput metab- disadvantage of NMR compared to mass spectrometry-based
olomics measurements, obtaining high spectral resolution techniques is somewhat overcome by the inherent capability of
remains a challenge because mismatch in magnetic suscepti- NMR to monitor isotopomer enrichment. The disadvantage of
bility of the sample and the chip material results in suscepti- mass spectrometry for measuring isotopomer distribution is
bility broadening. An innovative strategy to overcome based on spectral interpretation; the investigator must deduce
susceptibility broadening in chip-based NMR probes relies on from fragmentation patterns the location of enrichment,
the simulation of field maps to optimize the shape of air filled reducing sensitivity and throughput. The fate of labeled
compensating structures that ensure a flat magnetic field precursors (especially 13C labeled glycolytic intermediates)
distribution inside the sample detection region.65 has been tracked with NMR (or MRS) in vivo using hyper-
High-Resolution Magic Angle Spinning (HR-MAS). polarization and DNP. To better appreciate the versatility of
HR-MAS experiments provide 1H NMR spectra of semisolid NMR for exploring biological processes, the application of
materials such as cells, tissues, biopsy samples, organs, and NMR for metabolic flux measurements (including isotope
organisms. Spinning the sample about an axis at the magic tracing), isotope labeling, and DNP are discussed below.
angle (54°44′) reduces line broadening by averaging magnetic Isotope Labeling. Although NMR has inherent advantages
field gradients at compartment boundaries, the effects of for measuring in vivo isotopologue enrichment from 13C
residual homonuclear dipolar interactions, and chemical shift and 15N labeled substrates, it may still be difficult to identify
anisotropy.66 A complication in interpreting HR-MAS spectra and quantify enrichment. Many experiments permit the direct
is that metabolite chemical shifts can vary slightly according to detection of 13C to determine isotopomer distribution;71−74
the local microenvironment in tissues or cells (e.g., pH) ham- however, these experiments can still suffer from a lack of
pering accurate quantitation of metabolite concentrations. Lazariev sensitivity for isotopomers that are only partially enriched.
et al. describe an accurate method coupling quantum mechan- Although the enrichment of a given isotopomer may be minor,
ical simulations and quantitation algorithms (QM-QUEST) this change may still be important to understanding the
that corrects mismatches producing corrected metabolite finger- biological mechanisms of the system or pathway. Alternatively,
prints.67 1D 1H experiments can be used by taking advantage of the
An exciting advance in HR-MAS is the reduction in sample splitting in the 1H spectrum resulting from 13C enrichment,
size requirements using microcoil NMR probes. Wong et al. though 1H spectra at natural abundance can be convoluted
report the use of magic angle coil spinning (MACS) in which a even without the additional complication arising from the extra
62 μm diameter copper wire was wound around a quartz signals due to 13C couplings. To reduce the spectral complexity
capillary tube and soldered to a nonmagnetic capacitor arising from 13C enrichment, Cahoreau et al. used 2D
producing a detection volume of 690 nL.68 The capillary and heteronuclear J-resolved spectroscopy (2D-JRES), which
coil were fit inside a ceramic insert and placed in a standard records the scalar couplings in the second dimension instead
4 mm rotor. This setup allowed the measurement of 1H NMR of chemical shifts, decreasing experiment time compared with
spectra for a 500 ng sample of muscle tissue and produced a other 2D experiments.75 Other 2D experiments (such as
1
7-fold gain in sensitivity compared with a spectrum measured H−13C HSQC) can also be used to monitor isotopomer
for 16 mg of tissue packed into a 30 μL disposable insert, enrichment by taking advantage of the dispersion of a second
though the resonances in the MACS spectra were broader than dimension and the enhanced detection sensitivity of the 1H
desired for detailed metabolic phenotyping. Refinements of nucleus coupled to 13C.76 A recent publication by Fan and Lane
the initial MACS design were reported that minimize line applied a variety of 2D and 2D-edited experiments on either
broadening from temperature gradients and anisotropic [U−13C]-glucose or [U−13C, 15N]-glutamine enriched cancer
magnetic susceptibility to produce resonance line widths less cells.77 The 2D experiments described include 1H−1H TOCSY,
138 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

Figure 6. Workflow implemented in IsoDesign to determine the optimal label input in a 13C-MFA experiment. Blue, orange, and green boxes detail
steps performed by the calculation module, the visualization module, and a spreadsheet program, respectively. Reprinted from ref 92 with permission
from John Wiley & Sons Inc., copyright 2014.

1
H−1H HCCH TOCSY, 1H−13C HSQC, 1H−15N HSQC molecular biology and has recently been extensively re-
HACACO (correlating carbonyl carbons with neighboring viewed.82,83 The ultimate goal of MFA is to establish the
protons), 1H−13C HSQC-TOCSY, and 1H−31P HSQC metabolic flux through a particular pathway using labeled
TOCSY, and the authors discuss how the isotopomer distribution reagents and to compare the flux under different stresses and in
is determined from the spectra. Despite isotopic enrichment, various tissue types including cancers,84−86 oysters,87 plants,88
traditional 2D experiments can still be time-consuming and less and bacteria.89 These analyses are often limited to specific, well-
amenable to high-throughput isotope tracing than 1D experiments. defined biochemical pathways. Flux experiments measure time-
The advent of ultrafast 2D NMR experiments, which allow the based isotope enrichments and require that the activities of the
acquisition of a multidimensional NMR experiment in a single enzymes involved in the pathway are known. For well-studied
scan, has greatly reduced experiment times.78 Giraudeau and co- pathways, enzymatic activities can often be approximated;
workers measured the 13C enrichment in a biological sample with otherwise, parameters must be estimated to understand how
a traditional TOCSY experiment, which had a run time of greater the pathways may be affected. Regardless, computational
than 2 h, compared to an ultrafast TOCSY with an experiment methods are necessary to model the pathways and estimate
time of 3 s on a 400 MHz spectrometer equipped with a room the flux parameters. One such program reported by Hettling
temperature probe.58 Continued development and applications of and co-workers estimated TCA fluxes with noisy NMR data in
ultrafast 2D NMR experiments will significantly increase the heart tissue using a computational model to estimate the flux
throughput of isotope tracing by NMR. parameters.90 In addition to data analysis, the actual design of a
This section has predominantly discussed the use of isotope MFA study can be challenging, as one must consider what
labeling for the use of in vivo isotope labeling for the analysis of precursors and isotopic composition to use and the desired
biological pathways. Another interesting approach is to use outcome (dependent on the biological question under
isotope labeling ex vivo to improve NMR sensitivity for general investigation). Work by Millard et al. presents a novel software
metabolomics experiments. By incorporating an NMR active program for the design of 13C-MFA experiments, termed
isotope onto a tag, specific chemical moieties, such carboxylic IsoDesign.91 As illustrated in Figure 6, the program allows the
acid groups or amines, can be derivatized significantly investigator to generate models using different label inputs,
increasing sensitivity and selectivity while decreasing spectral simulate the isotopic data, calculate the precision expected on
convolution.79,80 For example, Tayyari and co-workers designed each flux for each label input, and outputs these results in
a cholamine tag with a 15N label on the primary amine, allowing a visually interpreted sensitivity landscape. Isodesign can be
derivatization of carboxylic acid functional groups.81 The used for comparison of isotopic data from MS (including
authors demonstrate the sensitivity of the tag using 1H−15N MS/MS) and NMR (both 1H and 13C). Tools such as those
HSQC experiments by collecting only 1−4 scans over 128 provided by Hettling et al., Millard et al., and others are
increments. The tag also included a tertiary amine which increasingly in need as more biological questions are being
provides a stable, permanent positive charge attached to all probed with substrates labeled specifically for the investigation
derivatized molecules, enhancing mass spectrometry sensitivity of selected pathways.90−92
as well. Although not yet widely used, the use of derivatization A major advantage of NMR over other popular MFA
to enhance NMR sensitivity and selectivity and decrease NMR analytical platforms is that it is nondestructive which allows for
resonance overlap while enhancing cross-platform utility has in vivo NMR spectroscopy under specific conditions. A
strong potential for improving NMR metabolite profiling comprehensive review and guide for in vivo NMR by de
experiments. Graaf and co-workers has recently been published and discusses
Metabolic Flux Analysis. Metabolic flux analysis (MFA) topics such as substrate choice, metabolic model, and choice of
is a popular and growing topic in analytical chemistry and NMR experiment (direct vs indirect detection).93 Although this
139 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

portion of the Review will briefly cover a few of the recently transferred to organic molecules by reacting hyperpolarized
published in vivo (or in situ) NMR experiments, the reader is hydrogen with precursor molecules in the presence of a
directed to the work by de Graaf and co-workers for a more in- transition metal catalyst. The use of PHIP for imaging,
depth discussion of the many considerations for in vivo 13C including the use of 13C or 15N labeled metabolite precursors,
NMR. Solid state NMR (ssNMR) can also be used for has been reviewed by Glöggler et al.99
MFA/isotope tracing. Work by Norris et al. on tree seedlings An exciting development in hyperpolarization experiments for
investigated both 13C and 15N allocation as a function of time, metabolic profiling is the rapidly expanding use of dynamic
allowing the researchers to monitor several different “pulses” of nuclear polarization (DNP), which has been accelerated by the
tracer and the spatial dispersion of the tracer before/after introduction of commercial instruments that facilitate its
administration.94 Although the high spin rates can result in implementation. An excellent overview of DNP in liquids has
detrimental effects on an in vivo system, it may provide a been recently authored by Günther.97 DNP takes advantage of
qualitative evaluation of substrate localization. Another the comparably larger polarization of electron spins, which can
approach that has been applied to study substrate localization, be nearly 100% at low temperatures. Saturation of the electron
while avoiding the physiological stress associated with high spin spin transfers this polarization from the unpaired electrons to
rates of ssNMR, is to use a “metabolomics-on-a-chip” method nuclear spin via hyperfine and dipolar interactions. Though
for MFA. Ouattara and co-workers recently reported the DNP experiments can be executed in a variety of experimental
application of a chip-based approach to explore the metabolic arrangements, the most useful configuration for metabolic
network of a hepatoma cell line.95 The authors compared the profiling experiments is dissolution DNP, in which polarization
chip environment with that of a traditional Petri dish and were is carried out at low temperatures (<1.5 K) and the polarized
able to determine that the cells in the chip showed evidence of liquid sample is generated by dissolution of the polarized solid
a higher oxygenation rate, indicating a more oxygen-rich and transferred to an NMR magnet for acquisition of the
environment. Although not necessarily an in vivo analysis, the NMR spectrum.
13
biochip method does allow for MFA to model biochemical C-labeled compounds are widely used in dissolution DNP
pathways and to deduce intracellular metabolite distributions in studies (in vitro and in vivo) because of their large chemical
controlled environments. shift range (∼200 ppm), low natural abundance (1.1%), which
Hyperpolarization and DNP. For much of its history, gives rise to a weak background, and the possibility of long
NMR has been considered to be a low sensitivity technique T1 relaxation times depending on the structural environ-
because small energy differences give rise to similar populations ment.100 DNP has been shown to increase the sensitivity of 13C
of the nuclear spin states at ambient temperatures. Hyper- detection by more than 10 000-fold enabling its use to probe
polarization is a method of increasing polarization by altering enzymatic processes in vivo.101 For example, DNP has been
the populations away from their thermal equilibrium value, used to follow the kinetics of pyruvate metabolism in cancer
enhancing sensitivity by several orders of magnitude. Because cells85,102 and in vivo in a breast cancer mouse model103 and to
the energy difference between two spin states is directly monitor the metabolism of U−13C2H7−glucose in perfused
proportional to the magnitude of the applied magnetic field, a human breast cancer cells.104 Schilling et al. demonstrated
simple (but expensive) mechanism for increasing polarization is the measurement of apparent diffusion coefficients using
to perform the measurements in a higher field magnet. hyperpolarized 13C lactate generated in vivo from [1-13C]-
However, the magnetic field required to reach a polarization pyruvate in tumor spheroids.105 Central carbon metabolism
of even 10% is outside the current technical limits. Another way has also been probed in Saccharomyces cerevisiae106,107 includ-
to increase polarization is by cooling to very low temperatures; ing an investigation of the effects of sulfite as an inhibitor of
for example, at 0.1 K, the polarization of protons reaches 23%.96 glycolysis.108
Though theoretically possible, measurements of NMR spectra Transfer of hyperpolarization to protons through indirect
of solid samples at such low temperatures would be quite detection experiments using spatially selective coherence trans-
challenging and the processes driving polarization buildup at fers offers a promising tool for implementation with standard
low temperatures become very slow. MRI scanners.109 As illustrated in Figure 7, the creative design
Hyperpolarization of various nuclei has proven to be a and application of hyperpolarized NMR probes can enable real-
powerful approach for enhancing the use of NMR for molecular time biological assays of probe uptake and export, pH, redox
and cellular imaging.97 For noble gases like 129Xe and 3He, state, reactive oxygen species, ion concentrations, signaling
hyperpolarization can be accomplished by optical pumping of pathways, etc.110 Hyperpolarized 89Y complexes are potentially
alkali metal atoms (typically Rb) which then transfer their attractive as NMR or MRI imaging probes because of the
polarization to the noble gas via spin exchange, increasing the unusually long T1 relaxation times (e.g., 10 min) typical of this
NMR signal by 10 000-fold. 129Xe gas has been used in many in very low γ nucleus. Lumata et al. demonstrate optimized
vivo imaging studies, particularly those targeting the lungs. DNP experiments leading to signal enhancements for 89Y over
Palaniappan et al. review the development of novel 129Xe 60 000 times the thermal signal.111
imaging agents composed of cryptophane cages coupled with a
biological recognition element like an antibody.98 Crypto-
phanes bind xenon with moderate affinity (∼4000 M−1), and
■ DATA PROCESSING AND CHEMOMETRICS
A significant challenge in metabolomics is the amount of data
residence times in the 30−300 ms range make them well-suited obtained from a single experiment. Even for NMR, which is
for in vivo MRI experiments. Another approach to hyper- inherently less sensitive than mass spectrometry, hundreds
polarization involves reactions of para-hydrogen. Gaseous of peaks are often detected and only a subset of these is iden-
hydrogen can exist in two nuclear spin isomers (ortho- and tifiable and quantifiable. For example, Bouatra et al. recently
para-). The concentration of the para-isomer can be increased published a report on the human urine metabolome identi-
by cooling the gas and inducing the conversion using a cata- fying some 179 peaks by NMR alone.11 Data processing
lyst. This para-hydrogen induced polarization (PHIP) can be and chemometrics analyses convert NMR spectral data into
140 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

Figure 7. Principle of biological assays using hyperpolarized NMR probes. Hyperpolarization is optimized ex situ, and the hyperpolarized probe or
label is added to a biomolecule, cell extracts, or living cells to conduct biological assays for detection inside an NMR spectrometer. Reprinted from
ref 110. Published under Creative Commons 4.0, copyright 2014.

Figure 8. Principal component analysis scores plots of (a) 16 metabolites fitted independently from round one; (b) same as for part (a) but with one
metabolite (threonine) and one outlying sample (QXB1) excluded; (c) 37 metabolites fitted on the basis of exemplar spectra in round two, with one
outlying sample (QXB1) excluded. Data were log transformed and mean centered. Reprinted from ref 112. Copyright 2011 American Chemical
Society.

information that can be used to address the particular biological pipeline for the preprocessing and chemometric analysis of
problem being interrogated. NMR data could help to reduce analyst-dependent variations.
Spectral Processing. Before the NMR data can be Such a pipeline would ideally include Fourier transformation,
interrogated for statistical changes, the raw data must be zero filling, apodization, phase correction, referencing, align-
processed, including baseline correction, phasing, apodization, ment, integration, and inclusion of various chemometric
referencing, integration, and normalization. This can be approaches (such as PCA, PLS, etc.). Recently, Worley and
accomplished manually, but manual data processing is time- Powers presented a package, termed MVAPACK, for the
consuming and can be less reliable than automated processing. complete handling of NMR data, from preprocessing, through
Because of the time associated with data processing, several alignment, integration, and chemometrics.113 Although not
groups have been working on improving and automating necessarily entirely automated, the package does allow for
different aspects (or the entirety) of the process. Data importing both Bruker and Agilent data through secondary
processing steps should be treated systematically to minimize programs.
introduction of errors. Tredwell et al. conducted a variability The individual aspects of the NMR preprocessing pipeline
study in which 5 different analysts independently processed have also been addressed. Improvements in addressing phase
the same data set.112 The five participants were asked to use correction,114 baseline correction,115 alignment,116−118 normal-
commercially available software (Chenomx NMR Suite) to fit ization,119,120 integration/binning,121 deconvolution,122−124 and
metabolite resonances in the same spectra of yeast extracts. quantitation125 have also been recently published. An interest-
Comparison of the results obtained by each analyst gave an ing approach reported by Romano et al. provides a program
overall coefficient of variation (CV) of 20%. In contrast, when PRICONA that uses the free induction decay (FID), rather
a single person performed the analysis of the same data than the processed spectrum for PCA analysis, and allevi-
5 times, the CV was only 2.4%. The variance between analysts ates the need for any operator manipulation.126 Continued
was largely attributed to differences in fitting spectral regions improvement and incorporation of emerging techniques for
having extensive peak overlap. Figure 8a shows PCA score NMR data preprocessing is essential for improving the high-
plots representing the 16 metabolites identified and quantified throughput characteristics of NMR for metabolite profiling.
by all 5 participants. Person 2 appears to be an outlier in Figure 9 provides examples of resonance deconvolution using
all samples, but with the removal of threonine and one out- an automated quantum mechanical total line shape model
lier sample (QXB1), Figure 8b shows better clustering.112 (QMTLS) for analysis of the NMR spectrum of a serum
Figure 8c shows the results of a second round of data sample treated by ultrafiltration (UF).127 Figure 9b highlights a
processing in which spectra were first processed by deconvoluted region in which the acetate resonance is over-
automation and then refit by each analyst, resulting in lapped with the multiplets of arginine and lysine. The quality
identification of 37 metabolites. The availability of a single of each fit was determined using relative-root-square-error
141 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

Quantitation. The popularity of quantitative NMR (qNMR)


has grown substantially over the past two decades. In
metabolomics and metabolic profiling experiments, qNMR can
provide absolute or relative quantification of multiple metabolites
within a sample without prior separation of components.135,136
While qNMR is a useful technique for metabolite quantification,
it is a demanding process with complications arising from water
suppression, T2-editing, protein interactions, relaxation differ-
ences, macromolecule background, and spectral overlap.137,138
To address these complications, quantification software
applications have been developed including MetaboQuant139
and IQMNMR.140 Several alternatives to quantitative 1D 1H
NMR spectra have been introduced, such as 2-D [1H−13C]
HSQC,141 2-D 1H INADEQUATE,142 and 1-D TOCSY.143
While 2-D [1H−13C] HSQC spectra offer the benefit of
reduced spectral overlap and improved resolution, metabo-
lite quantification can be compromised by resonance-specific
signal attenuation during coherence transfer delays.141 Hu
et al. demonstrated that, for accurate quantitation, a series of
HSQC spectra can be acquired with incremented repetition
times. Back extrapolation of the data yields a time-zero HSQC
spectrum in which metabolite resonance intensities are directly
proportional to concentrations.141 Because their acquisition
can be time-consuming, these 2D-experiments benefit from the
use of cryogenic probes, which increase measurement
sensitivity by reducing noise and thereby decreasing time
spent signal averaging or by the use of fast/ultrafast NMR
experiments.
Figure 9. Examples of deconvolution with doublets of valine, isoleucine,
Chemometrics. Characterization of the processed NMR
and ketoleucine and triplets of leucine and isoleucine (A) and singlet of
acetate and complex multiplets of arginine and lysine (B) of a single UF data is another important facet of the NMR metabolomics
serum sample. Reprinted from ref 127 with kind permission from pipeline. With hundreds of resonances possible in a biological
Springer Science and Business Media, copyright 2014. sample, making sense of the peak identities and intensity
changes can be challenging. Such analyses are often performed
using complicated algorithms and combining several chemo-
(RRMSE), and a good fit was characterized by a low RRMSE metric approaches to indicate the important resonances prior to
value as well as a high R factor. metabolite identification. Several different approaches were
Resonance Assignments and Databases. One of the evaluated for their ability to facilitate classification using five
major challenges of NMR metabolic profiling is the ability to biofluid 1H NMR data sets, including urine, plasma, milk, and
make resonance assignments from the obtained spectral data. serum.144 The methods interrogated by Hochrein et al. include
Often, 1H NMR spectra have a high degree of spectral overlap Support Vector Machines, Elastic Net, Random Forests, Top
making assignments challenging.128 Peak assignments in these Scoring Pairs, and Nearest Shrunken Centroids. The authors
cases can be made with the help of 2D HSQC spectra. found that, unsurprisingly, many methods were dependent on
The added dispersion in the 13C dimension improves the the data set being investigated. Only Support Vector Machines
resolution of peaks overlapped in the 1H spectrum and adds and Random Forests combined with other statistical ap-
13
C chemical shifts as an additional parameter to aid in proaches for filtering gave the best performance for a majority
resonance assignments.128 of the data sets.144 Similarly, Maraschin et al. investigated the
While multidimensional NMR experiments can aid in the combination of several different methods to classify the
process of assigning resonances, this strategy can be time- geographical origin of a native Brazilian plant and found that
consuming.129 When the identity of the metabolites in a sample a combination of several algorithms was optimal.145 Other
are known (or suspected), resonance assignments can be advances in chemometrics include the application of techniques
facilitated using libraries or databases of 1H NMR spectra such specifically for certain NMR experiments in which multidimen-
as HMDB and MMCD.130 These databases typically display sional data sets (such as two-dimensional J-resolved spectros-
spectra as static images which do not allow for detailed copy) are decomposed into matrices that describe the different
inspection of a particular region,131 lack the ability to perform components of the 2D experiment (J-couplings, chemical shift,
automated assignments, and can be time-consuming to and intensities).124 The resultant matrices can then be
search.132 While databases commonly provide lists of chemi- subjected to traditional multivariate statistical analysis with-
cal shifts for each metabolite, they may lack information about out sacrificing the additional information provided by the
coupling constants. Coupling constants provide information 2D experiment.
about atom connectivity, stereochemistry, and molecular con- Other methods for reducing the complexity of NMR data
formation making them useful for resonance assignments, include the statistical integration of NMR data from different
especially in complex spectra.131 Many investigators have begun biofluids/tissues from the same specimen,146 removal of inter-
developing their own databases including MetaboID,129 fering resonances from pharmaceuticals,147 or high concen-
Metabohunter,130 MetIDB,131 MeRy-B,133 and BML-NMR.134 trations of metabolites, such as sugars in diabetic patients that
142 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

Figure 10. Comparison of STOCSY and STORM for the identification of N-methylnicotinic acid signals in the INTERMAP BMI nondiabetic U.S.
data set. Scatterplot of correlations determined by STOCSY (x-axis, n = 1880) and by STORM (y-axis, n = 112), using an N-methylnicotinic acid
(NMNA) peak at δ 8.84 as driver, is shown. For each axis, the kernel density estimation of the distribution of each class is shown in the same color
on the opposite side. NMNA is shown in orange, hippuric acid in green, an unknown metabolite in light blue, and all other variables in dark blue.
For each of the metabolites, each signal is shown with a different symbol. For clarity, each distribution has been normalized to the same total area.
It shows that the density estimate of NMNA overlaps less with others in STORM compared to STOCSY. Reprinted from ref 149. Copyright 2012
American Chemical Society.

might be obscuring other resonances.123 Wei et al. addresses spectra measured for a collection of samples from a general
the problem of identifying resonances in a complex sample population. Additional approaches for identifying and quantify-
using ratio analysis NMR spectroscopy (RANSY).128 Like ing substituents in complex samples have also been published
STOCSY (statistical total correlation spectroscopy),148 RANSY or reviewed151−153 and are of continuing interest to the
is a statistical approach centered on identifying resonances bioanalytical community.
of the same molecule from a series of spectra, though it does
not rely on statistical correlations. Instead with RANSY, the
ratios of the detected peaks are used to determine molecular
■ PERSPECTIVE
NMR-based metabolomics has rapidly become established as a
connectivity. Recent variations of STOCSY have also been useful approach for the analysis of complex biosamples
reported including iterative STOCSY (I-STOCSY)132 and contributing to biomarker discovery, disease diagnosis, and
STORM (subset optimization by reference matching).149 treatment monitoring as well as providing insights into the
STORM provides subset optimization by reference matching impact of environmental stressors on organisms and eco-
using a 3-step algorithm that includes subset selection, systems. In addition to untargeted metabolomics measure-
STOCSY of the subset, and reference updating. One downfall ments, isotopic labeling and metabolic flux experiments provide
of STOCSY is difficulty in identifying low concentration metab- unique insights into specific metabolic pathways. Innovations
olites, but Posma et al. posit that this is improved in STORM related to automated data processing, resonance assignments,
through the addition of subset selection.149 Figure 10 provides and statistical data analysis continue to improve the reliability
a comparison of STORM (y-axis) and STOCSY (x-axis) for and robustness of NMR metabolomics results. The introduc-
resonances in a large data set of urine samples from nondiabetic tion of fast and ultrafast multidimensional NMR experiments
patients.149 Note that the N-methylnicotinic acid (NMNA) promises to greatly improve the throughput of NMR
density estimate, shown in orange, has less overlap with other measurements. In addition, approaches that affect nuclear
components using STORM, making it possible to recover rare polarization such as DNP dramatically enhance NMR
signals obscured by resonance overlap. Another promising sensitivity. Rather than choosing between NMR and mass
technique for biomarker identification is visual interpretation of spectrometry-based metabolomics, strategies that make use
z-score ratios (VIZR) developed by Barding et al.150 VIZR of both platforms provide better coverage of the metabolome
identifies unique resonances using ratios of z-scores calculated and improved measurement reliability. Chemometric tools
by comparing a spectrum obtained for a subject sample to that integrate the results of NMR, GC/MS, and LC-MS and
143 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146
Analytical Chemistry Review

results from other analytical techniques will benefit the adop- (7) Duarte, I. F.; Diaz, S. O.; Gil, A. M. J. Pharm. Biomed. Anal. 2014,
tion of multiplatform metabolomics approaches. 93, 17−26.


(8) Duarte, I. F.; Gil, A. M. Prog. Nucl. Magn. Reson. Spectrosc. 2012,
62, 51−74.
AUTHOR INFORMATION
(9) Hyndman, M. E.; Mullins, J. K.; Bivalacqua, T. J. Urol. Oncol.:
Corresponding Author Semin. Orig. Invest. 2011, 29, 558−561.
*E-mail: clarive@ucr.edu. (10) Nicholson, G.; Rantalainen, M.; Maher, A. D.; Li, J. V.;
Malmodin, D.; Ahmadi, K. R.; Faber, J. H.; Hallgrimsdottir, I. B.;
Notes
Barrett, A.; Toft, H.; Krestyaninova, M.; Viksna, J.; Neogi, S. G.;
The authors declare no competing financial interest. Dumas, M. E.; Sarkans, U.; Silverman, B. W.; Donnelly, P.; Nicholson,
Biographies J. K.; Allen, M.; Zondervan, K. T.; Lindon, J. C.; Spector, T. D.;
Cynthia K. Larive is a Professor of Chemistry at the University of McCarthy, M. I.; Holmes, E.; Baunsgaard, D.; Holmes, C. C. Mol. Syst.
California−Riverside (UCR) and currently serves as Divisional Dean Biol. 2011, 7, 525.
(11) Bouatra, S.; Aziat, F.; Mandal, R.; Guo, A. C.; Wilson, M. R.;
within the College of Natural and Agricultural Sciences. She received
Knox, C.; Bjorndahl, T. C.; Krishnamurthy, R.; Saleem, F.; Liu, P.;
her B.S. degree from South Dakota State University in 1980 and a
Dame, Z. T.; Poelzer, J.; Huynh, J.; Yallou, F. S.; Psychogios, N.;
Ph.D. from UCR in 1992 under the direction of Dallas Rabenstein. Dong, E.; Bogumil, R.; Roehring, C.; Wishart, D. S. PLoS One 2013, 8,
She was on the faculty of the University of Kansas from 1992 to 2004, No. e73076.
before returning to UCR in 2005. Larive is a fellow of the AAAS and (12) Scano, P.; Locci, E.; Noto, A.; Navarra, G.; Murgia, F.; Lussu,
ACS and has received awards for teaching, research, and service. Her M.; Barberini, L.; Atzori, L.; De Giorgio, F.; Rosa, M. F.; d’Aloja, E.
research interests are in the application of NMR, GC/MS and LC-MS Magn. Reson. Chem. 2013, 51, 454−462.
for metabolomics, and metabolic profiling in a variety of biological (13) Izquierdo-García, J. L.; Nin, N.; Ruíz-Cabello, J.; Rojas, Y.; de
systems and in the structural characterization of the glycosaminogly- Paula, M.; López-Cuenca, S.; Morales, L.; Martínez-Caro, L.;
cans heparin and heparan sulfate. Fernández-Segoviano, P.; Esteban, A.; Lorente, J. A. Intensive Care
Med. 2011, 37, 2023−2032.
Gregory A. Barding, Jr. is an Assistant Professor of Chemistry at
(14) DeFeo, E. M.; Wu, C. L.; McDougal, W. S.; Cheng, L. L. Nat.
California Polytechnic University, Pomona. He received his B.S. Rev. Urol. 2011, 8, 301−311.
degree from California State University−San Bernardino in 2008 and (15) Zhang, B.; Halouska, S.; Schiaffo, C. E.; Sadykov, M. R.;
his Ph.D. degree in 2013 from UCR where he was mentored jointly by Somerville, G. A.; Powers, R. J. Proteome Res. 2011, 10, 3743−3754.
Cynthia Larive and Julia Bailey-Serres in the Department of Botany (16) Garcia-Á lvarez, I.; Fernández-Mayoralas, A.; Garrido, L. Curr.
and Plant Sciences. He was a postdoctoral scientist at the University of Top. Med. Chem. 2011, 11, 27−42.
Washington School of Medicine in the laboratory of professor Daniel (17) Duarte, I. F. J. Controlled Release 2011, 153, 34−39.
Raftery from 2013 to 2014. Dr. Barding’s research interests are focused (18) Rolin, D.; Deborde, C.; Maucourt, M.; Cabasson, C.; Fauvelle,
on applying various analytical techniques including GC, LC, NMR, F.; Jacob, D.; Canlet, C.; Moing, A. In Metabolomics Coming of Age with
UV−vis, and mass spectrometry for general metabolite profiling Its Technological Diversity; Rolin, D., Ed; Adv. Bot. Res. 67; Academic
studies as well as stable isotope tracing in several biological systems. Press: London, UK, 2013; pp 1−66.
(19) Barding, G. A., Jr.; Fukao, T.; Beni, S.; Bailey-Serres, J.; Larive,
Meredith M. Dinges is a Ph.D. candidate at UCR under the mentorship C. K. J. Proteome Res. 2012, 11, 337−347.
of Cynthia Larive. She received her B.A. degree from Drury University (20) Barding, G. A., Jr.; Fukao, T.; Beni, S.; Bailey-Serres, J.; Larive,
in 2012. Her research interests are focused on metabolomics and C. K. J. Proteome Res. 2013, 12, 898−909.
metabolic profiling of fecal samples along the length of the colon to (21) Orr, D. J.; Barding, G. A.; Tolley, C. E.; Hicks, G. R.; Raikhel, N.
understand the dynamic processes related to microbial metabolism V.; Larive, C. K. In Plant Chemical Genomics: Methods and Protocols;
and transport of metabolites across the lumen and into the Hicks, G. R., Robert, S., Eds.; Methods Mol. Biol. 1056; Springer: New
bloodstream. She carries out her research in collaboration with York, USA, 2014; pp 225−239.
Professor Christian Lytle in the UCR School of Medicine. (22) Kim, H. K.; Choi, Y. H.; Verpoorte, R. Trends Biotechnol. 2011,


29, 267−275.
(23) Serra, O.; Chatterjee, S.; Huang, W. L.; Stark, R. E. Plant Sci.
ACKNOWLEDGMENTS 2012, 195, 120−124.
Support by the National Science Foundation (Grant No. IOS- (24) Leiss, K. A.; Choi, Y. H.; Verpoorte, R.; Klinkhamer, P. G. L.
1121626) and the U.S. Department of Agriculture, National Phytochem. Rev. 2011, 10, 205−216.
Institute of Food and Agriculture - Agriculture and Food (25) Forseth, R. R.; Schroeder, F. C. Curr. Opin. Chem. Biol. 2011, 15,
38−47.
Research Initiative (Grant No. 2011−04015) to C.K.L. is
(26) Robinette, S. L.; Bruschweiler, R.; Schroeder, F. C.; Edison, A. S.
gratefully acknowledged.


Acc. Chem. Res. 2012, 45, 288−297.
(27) Lankadurai, B. P.; Nagato, E. G.; Simpson, M. J. Environ. Rev.
REFERENCES 2013, 21, 180−205.
(1) Dunn, W. B.; Broadhurst, D. I.; Atherton, H. J.; Goodacre, R.; (28) Ogata, Y.; Chikayama, E.; Morioka, Y.; Everroad, R. C.; Shino,
Griffin, J. L. Chem. Soc. Rev. 2011, 40, 387−426. A.; Matsushima, A.; Haruna, H.; Moriya, S.; Toyoda, T.; Kikuchi, J.
(2) Robinette, S. L.; Holmes, E.; Nicholson, J. K.; Dumas, M. E. PLoS One 2012, 7, No. e30263.
Genome Med. 2012, 4, 30. (29) Heyman, H. M.; Meyer, J. J. M. S. Afr. J. Bot. 2012, 82, 21−32.
(3) Bothwell, J. H. F.; Griffin, J. L. Biol. Rev. 2011, 86, 493−510. (30) Hong, Y. S. Magn. Reson. Chem. 2011, 49, S13−S21.
(4) Gebregiworgis, T.; Powers, R. Comb. Chem. High Throughput (31) Abriata, L. A. Concepts Magn. Reson., Part A 2012, 40A, 171−
Screening 2012, 15, 595−610. 178.
(5) Emwas, A.-H. M.; Salek, R. M.; Griffin, J. L.; Merzaban, J. (32) Zhang, A. H.; Sun, H.; Qiu, S.; Wang, X. J. Magn. Reson. Chem.
Metabolomics 2013, 9, 1048−1072. 2013, 51, 549−556.
(6) Mikkonen, J. J. W.; Herrala, M.; Soininen, P.; Lappalainen, R.; (33) Keun, H. C.; Athersuch, T. J. In Metabolic Profiling: Methods and
Tjaderhane, L.; Seitsalo, H.; Niemela, R.; Salo, T.; Kullaa, A. M.; Protocols, Metz, T. O., Ed.; Methods Mol. Biol. 708; Springer: New
Myllymaa, S. Metabolomics 2013, 3, 128. York, USA, 2011; pp 321−334.

144 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146


Analytical Chemistry Review

(34) Lacy, P.; McKay, R. T.; Finkel, M.; Karnovsky, A.; Woehler, S.; (66) Hu, J. Z. In Metabolic Profiling: Methods and Protocols; Metz, T.
Lewis, M. J.; Chang, D.; Stringer, K. A. PLoS One 2014, 9, No. e85732. O., Ed.; Methods Mol. Biol. 708; Springer: New York, USA, 2011; pp
(35) Simon-Manso, Y.; Lowenthal, M. S.; Kilpatrick, L. E.; Sampson, 335−364.
M. L.; Telu, K. H.; Rudnick, P. A.; Mallard, W. G.; Bearden, D. W.; (67) Lazariev, A.; Allouche, A. R.; Aubert-Frecon, M.; Fauvelle, F.;
Schock, T. B.; Tchekhovskoi, D. V.; Blonder, N.; Yan, X. J.; Liang, Y. Piotto, M.; Elbayed, K.; Namer, I. J.; van Ormondt, D.; Graveron-
X.; Zheng, Y. F.; Wallace, W. E.; Neta, P.; Phinney, K. W.; Remaley, A. Demilly, D. Meas. Sci. Technol. 2011, 22, 114020.
T.; Stein, S. E. Anal. Chem. 2013, 85, 11725−11731. (68) Wong, A.; Jiménez, B.; Li, X. N.; Holmes, E.; Nicholson, J. K.;
(36) Rasmussen, L. G.; Savorani, F.; Larsen, T. M.; Dragsted, L. O.; Lindon, J. C.; Sakellariou, D. Anal. Chem. 2012, 84, 3843−3848.
Astrup, A.; Engelsen, S. B. Metabolomics 2011, 7, 71−83. (69) Wong, A.; Li, X. N.; Sakellariou, D. Anal. Chem. 2013, 85,
(37) Pinto, J.; Domingues, M. R. M.; Galhano, E.; Pita, C.; Almeida, 2021−2026.
M. D.; Carreira, I. M.; Gil, A. M. Analyst 2014, 139, 1168−1177. (70) Wong, A.; Li, X. N.; Molin, L.; Solari, F.; Elena-Herrmann, B.;
(38) Gowda, G. A. N.; Raftery, D. Anal. Chem. 2014, 86, 5433−5440. Sakellariou, D. Anal. Chem. 2014, 86, 6064−6070.
(39) Jiang, L. M.; Huang, J.; Wang, Y. L.; Tang, H. R. Analyst 2012, (71) El Hage, M.; Conjard-Duplany, A.; Baverel, G.; Martin, G.
137, 4209−4219. Neurochem. Int. 2011, 58, 896−903.
(40) Matheus, N.; Hansen, S.; Rozet, E.; Peixoto, P.; Maquoi, E.; (72) Gonzalez-Rodriguez, I.; Gaspar, P.; Sanchez, B.; Gueimonde,
Lambert, V.; Noël, A.; Frédérich, M.; Mottet, D.; de Tullio, P. M.; Margolles, A.; Neves, A. R. Appl. Environ. Microb. 2013, 79, 7628−
Phytochem. Anal. 2014, 25, 342−349. 7638.
(41) Martineau, E.; Tea, I.; Loaec, G.; Giraudeau, P.; Akoka, S. Anal. (73) Li, W.; Bian, F.; Chaudhuri, P.; Mao, X.; Brunengraber, H.; Yu,
Bioanal. Chem. 2011, 401, 2133−2142. X. NMR Biomed. 2011, 24, 176−187.
(42) Jacobs, D. M.; Spiesser, L.; Garnier, M.; de Roo, N.; van (74) Marin-Valencia, I.; Cho, S. K.; Rakheja, D.; Hatanpaa, K. J.;
Dorsten, F.; Hollebrands, B.; van Velzen, E.; Draijer, R.; van Kapur, P.; Mashimo, T.; Jindal, A.; Vemireddy, V.; Good, L. B.;
Duynhoven, J. Anal. Bioanal. Chem. 2012, 404, 2349−2361. Raisanen, J.; Sun, X.; Mickey, B.; Choi, C.; Takahashi, M.; Togao, O.;
(43) van der Hooft, J. J. J.; de Vos, R. C. H.; Mihaleva, V.; Bino, R. J.; Pascual, J. M.; DeBerardinis, R. J.; Maher, E. A.; Malloy, C. R.; Bachoo,
Ridder, L.; de Roo, N.; Jacobs, D. M.; van Duynhoven, J. P. M.; R. M. NMR Biomed. 2012, 25, 1177−1186.
Vervoort, J. Anal. Chem. 2012, 84, 7263−7271. (75) Cahoreau, E.; Peyriga, L.; Hubert, J.; Bringaud, F.; Massou, S.;
(44) van der Hooft, J. J. J.; Mihaleva, V.; de Vos, R. C. H.; Bino, R. J.; Portais, J.-C. Anal. Biochem. 2012, 427, 158−163.
Vervoort, J. Magn. Reson. Chem. 2011, 49, S55−S60. (76) Gallinger, A.; Biet, T.; Pellerin, L.; Peters, T. Angew. Chem., Int.
(45) van der Hooft, J. J. J.; de Vos, R. C. H.; Ridder, L.; Vervoort, J.; Ed. 2011, 50, 11672−11674.
Bino, R. J. Metabolomics 2013, 9, 1009−1018. (77) Fan, T. W.-M.; Lane, A.N. J. Biomol. NMR 2011, 49, 267−280.
(46) McKay, R. T. Concepts Magn. Reson., Part A 2011, 38A, 197− (78) Frydman, L.; Scherf, T.; Lupulescu, A. Proc. Natl. Acad. Sci.
220. U.S.A. 2002, 99, 15858−15862.
(47) Farooq, H.; Soong, R.; Courtier-Murias, D.; Anklin, C.; (79) Shanaiah, N.; Desilva, M. A.; Gowda, G. A. N.; Raftery, M. A.;
Simpson, A. Anal. Chem. 2012, 84, 6759−6766.
Hainline, B. E.; Raftery, D. Proc. Natl. Acad. Sci. U.S.A. 2007, 104,
(48) Martin-Pastor, M. J. Agric. Food Chem. 2014, 62, 1190−1197.
11540−11544.
(49) Huang, Y. Q.; Cal, S. H.; Zhang, Z. Y.; Chen, Z. Biophys. J. 2014,
(80) Ye, T.; Mo, H.; Shanaiah, N.; Gowda, G. A. N.; Zhang, S.;
106, 2061−2070.
Raftery, D. Anal. Chem. 2009, 81, 4882−4888.
(50) Huang, Y. Q.; Zhang, Z. Y.; Cai, S. H.; Chen, Z. PLoS One 2013,
(81) Tayyari, F.; Gowda, G. A. N.; Gu, H. W.; Raftery, D. Anal.
9, No. e84032.
(51) Shemesh, N.; Dumez, J. N.; Frydman, L. Chem.Eur. J. 2013, Chem. 2013, 85, 8715−8721.
(82) Sims, J. K.; Manteiga, S.; Lee, K. Curr. Opin. Biotechnol. 2013,
19, 13002−13008.
(52) Hoch, J. C.; Maciejewski, M. W.; Mobli, M.; Schuyler, A. D.; 24, 933−939.
Stern, A. S. Acc. Chem. Res. 2014, 47, 708−717. (83) Fernie, A. R.; Morgan, J. A. Plant Cell Environ. 2013, 36, 1738−
(53) Rai, R. K.; Sinha, N. Anal. Chem. 2012, 84, 10005−10011. 1750.
(54) Pathan, M.; Akoka, S.; Tea, I.; Charrier, B.; Giraudeau, P. (84) Yang, Y.; Lane, A. N.; Ricketts, C. J.; Sourbier, C.; Wei, M.-H.;
Analyst 2011, 136, 3157−3163. Shuch, B.; Pike, L.; Wu, M.; Rouault, T. A.; Boros, L. G.; Fan, T. W.
(55) Le Guennec, A.; Giraudeau, P.; Caldarelli, S. Anal. Chem. 2014, M.; Linehan, W. M. PLoS One 2013, 8, No. e72179.
86, 5946−5954. (85) Yang, C.; Harrison, C.; Jin, E. S.; Chuang, D. T.; Sherry, A. D.;
(56) Pathan, M.; Akoka, S.; Giraudeau, P. J. Magn. Reson. 2012, 214, Malloy, C. R.; Merritt, M. E.; DeBerardinis, R. J. J. Biol. Chem. 2014,
335−339. 289, 6212−6224.
(57) Giraudeau, P.; Cahoreau, E.; Massou, S.; Pathan, M.; Portais, J. (86) Cheng, T.; Sudderth, J.; Yang, C.; Mullen, A. R.; Jin, E. S.;
C.; Akoka, S. ChemPhysChem 2012, 13, 3098−3101. Mates, J. M.; DeBerardinis, R. J. Proc. Natl. Acad. Sci. U.S.A. 2011, 108,
(58) Giraudeau, P.; Massou, S.; Robin, Y.; Cahoreau, E.; Portais, J.- 8674−8679.
C.; Akoka, S. Anal. Chem. 2011, 83, 3112−3119. (87) Tikunov, A. P.; Stoskopf, M. K.; Macdonald, J. M. Metabolites
(59) Giraudeau, P.; Akoka, S. In Metabolomics Coming of Age with Its 2014, 4, 53−70.
Technological Diversity; Rolin, D., Ed; Adv. Bot. Res. 67; Academic (88) Gilbert, A.; Robins, R. J.; Remaud, G. S.; Tcherkez, G. G. B.
Press: London, UK, 2013; pp 99−158. Proc. Natl. Acad. Sci. U.S.A. 2012, 109, 18204−18209.
(60) Renslow, R. S.; Babauta, J. T.; Majors, P. D.; Mehta, H. S.; (89) Rados, D.; Turner, D. L.; Fonseca, L. L.; Carvalho, A. L.;
Ewing, R. J.; Ewing, T. W.; Mueller, K. T.; Beyenal, H. Water Sci. Blombach, B.; Eikmanns, B. J.; Neves, A. R.; Santos, H. Appl. Environ.
Technol. 2014, 69, 966−973. Microbiol. 2014, 80, 3015−3024.
(61) Ramaswamy, V.; Hooker, J. W.; Withers, R. S.; Nast, R. E.; Brey, (90) Hettling, H.; Alders, D. J. C.; Heringa, J.; Binsl, T. W.;
W. W.; Edison, A. S. J. Magn. Reson. 2013, 235, 58−65. Groeneveld, A. B. J.; van Beek, J. H. G. M. BMC Syst. Biol. 2013, 7, 82.
(62) Tadanki, S.; Colon, R. D.; Moore, J.; Waddell, K. W. J. Magn. (91) Millard, P.; Sokol, S.; Letisse, F.; Portais, J.-C. Biotechnol. Bioeng.
Reson. 2012, 223, 64−67. 2014, 111, 202−208.
(63) Grimes, J. H.; O’Connell, T. M. J. Biomol. NMR 2011, 49, 297− (92) Moseley, H. N. B.; Lane, A. N.; Belshoff, A. C.; Higashi, R. M.;
305. Fan, T. W. M. BMC Biol. 2011, 9, 37.
(64) Falck, D.; Oosthoek-de Vries, A. J.; Kolkman, A.; Lingeman, H.; (93) de Graaf, R. A.; Rothman, D. L.; Behar, K. L. NMR Biomed.
Honing, M.; Wijmenga, S. S.; Kentgens, A. P. M.; Niessen, W. M. A. 2011, 24, 958−972.
Anal. Bioanal. Chem. 2013, 405, 6711−6720. (94) Norris, C. E.; Quideau, S. A.; Landhausser, S. M.; Bernard, G.
(65) Ryan, H.; Smith, A.; Utz, M. Lab Chip 2014, 14, 1678−1685. M.; Wasylishen, R. E. Sci. Rep. 2012, 2, 719.

145 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146


Analytical Chemistry Review

(95) Ouattara, D. A.; Prot, J.-M.; Bunescu, A.; Dumas, M.-E.; Elena- (128) Wei, S. W.; Zhang, J.; Liu, L. Y.; Ye, T.; Gowda, G. A. N.;
Herrmann, B.; Leclerc, E.; Brochot, C. Mol. BioSyst. 2012, 8, 1908− Tayyari, F.; Raftery, D. Anal. Chem. 2011, 83, 7616−7623.
1920. (129) MacKinnon, N.; Somashekar, B. S.; Tripathi, P.; Ge, W. C.;
(96) Günther, U. L. Top. Curr. Chem. 2013, 335, 23−69. Rajendiran, T. M.; Chinnaiyan, A. M.; Ramamoorthy, A. J. Magn.
(97) Witte, C.; Schröder, L. NMR Biomed. 2013, 26, 788−802. Reson. 2013, 226, 93−99.
(98) Palaniappan, K. K.; Francis, M. B.; Pines, A.; Wemmer, D. E. Isr. (130) Tulpan, D.; Leger, S.; Belliveau, L.; Culf, A.; Cuperlovic-Culf,
J. Chem. 2014, 54, 104−112. M. BMC Bioinf. 2011, 12, 400.
(99) Gloggler, S.; Colell, J.; Appelt, S. J. Magn. Reson. 2013, 235, (131) Mihaleva, V. V.; te Beek, T. A. H.; van Zimmeren, F.; Moco, S.;
130−142. Laatikainen, R.; Niemitz, M.; Korhonen, S. P.; van Driel, M. A.;
(100) Keshari, K. R.; Wilson, D. M. Chem. Soc. Rev. 2014, 43, 1627− Vervoort, J. Anal. Chem. 2013, 85, 8700−8707.
1659. (132) Sands, C. J.; Coen, M.; Ebbels, T. M. D.; Holmes, E.; Lindon, J.
(101) Koptyug, I. V. Mendeleev Commun. 2013, 23, 299−312. C.; Nicholson, J. K. Anal. Chem. 2011, 83, 2075−2082.
(102) Harrison, C.; Yang, C.; Jindal, A.; DeBerardinis, R. J.; (133) Ferry-Dumazet, H.; Gil, L.; Deborde, C.; Moing, A.; Bernillon,
Hooshyar, M. A.; Merritt, M.; Sherry, A. D.; Malloy, C. R. NMR S.; Rolin, D.; Nikolski, M.; de Daruvar, A.; Jacob, D. BMC Plant Biol.
Biomed. 2012, 25, 1286−1294. 2011, 11, 104.
(103) Li, L. Z.; Kadlececk, S.; Xu, H. N.; Daye, D.; Pullinger, B.; (134) Ludwig, C.; Easton, J. M.; Lodi, A.; Tiziani, S.; Manzoor, S. E.;
Profka, H.; Chodosh, L.; Rizi, R. NMR Biomed. 2013, 26, 1308−1320. Southam, A. D.; Byrne, J. J.; Bishop, L. M.; He, S.; Arvanitis, T. N.;
(104) Harris, T.; Degani, H.; Frydman, L. NMR Biomed. 2013, 26, Gunther, U. L.; Viant, M. R. Metabolomics 2012, 8, 8−18.
1831−1843. (135) Bharti, S. K.; Roy, R. TrAC, Trends Anal. Chem. 2012, 35, 5−
(105) Schilling, F.; Düwel, S.; Köllisch, U.; Durst, M.; Schulte, R. F.; 26.
(136) Barding, G. A., Jr.; Salditos, R.; Larive, C. K. Anal. Bioanal.
Glaser, S.; Haase, A.; Otto, A. M.; Menzel, M. I. NMR Biomed. 2013,
Chem. 2012, 404, 1165−1179.
26, 557−568.
(137) Sokolenko, S.; Blondeel, E. J. M.; Azlah, N.; George, B.;
(106) Jensen, P. R.; Karlsson, M.; Lerche, M. H.; Meier, S. Chem.
Schulze, S.; Chang, D.; Aucoin, M. G. Anal. Chem. 2014, 86, 3330−
Eur. J. 2013, 19, 13288−13293.
3337.
(107) Meier, S.; Karlsson, M.; Jensen, P. R.; Lerche, M. H.; Duus, J. (138) Tiainen, M.; Soininen, P.; Laatikainen, R. J. Magn. Reson. 2014,
O. Mol. Biosyst. 2011, 7, 2834−2836. 242, 67−78.
(108) Meier, S.; Solodovnikova, N.; Jensen, P. R.; Wendland, J. (139) Klein, M. S.; Oefner, P. J.; Gronwald, W. BioTechniques 2013,
ChemBioChem 2012, 13, 2265−2269. 54, 251−256.
(109) Harris, T.; Giraudeau, P.; Frydman, L. Chem.Eur. J. 2011, 17, (140) Song, X.; Zhang, B. L.; Liu, H. M.; Yu, B. Y.; Gao, X. M.; Kang,
697−703. L. Y. BMC Bioinformatics 2011, 12, 337.
(110) Meier, S.; Jensen, P. R.; Karlsson, M.; Lerche, M. H. Sensors (141) Hu, K. F.; Westler, W. M.; Markley, J. L. J. Am. Chem. Soc.
2014, 14, 1576−1597. 2011, 133, 1662−1665.
(111) Lumata, L.; Jindal, A. K.; Merritt, M. E.; Malloy, C. R.; Sherry, (142) Martineau, E.; Giraudeau, P.; Tea, I.; Akoka, S. J. Pharm.
A. D.; Kovacs, Z. J. Am. Chem. Soc. 2011, 133, 8673−8680. Biomed. Anal. 2011, 54, 252−257.
(112) Tredwell, G. D.; Behrends, V.; Geier, F. M.; Liebeke, M.; (143) Sandusky, P.; Appiah-Amponsah, E.; Raftery, D. J. Biomol.
Bundy, J. G. Anal. Chem. 2011, 83, 8683−8687. NMR 2011, 49, 281−290.
(113) Worley, B.; Powers, R. ACS Chem. Biol. 2014, 9, 1138−44. (144) Hochrein, J.; Klein, M. S.; Zacharias, H. U.; Li, J.; Wijffels, G.;
(114) Worley, B.; Powers, R. Chemom. Intell. Lab. Syst. 2014, 131, 1− Schirra, H. J.; Spang, R.; Oefner, P. J.; Gronwald, W. J. Proteome Res.
6. 2012, 11, 6242−6251.
(115) Wang, K. C.; Wang, S. Y.; Kuo, C. H.; Tseng, Y. F. J. Anal. (145) Maraschin, M.; Somensi-Zeggio, A.; Oliveira, S. K.; Kuhnen, S.;
Chem. 2013, 85, 1231−1239. Tomazzoli, M. M.; Zeri, A. C. M.; Carreira, R.; Rocha, M. Lect. Notes
(116) Liebeke, M.; Hao, J.; Ebbels, T. M. D.; Bundy, J. G. Anal. Bioinf. 2012, 7632, 129−140.
Chem. 2013, 85, 4605−4612. (146) Maher, A. D.; Cysique, L. A.; Brew, B. J.; Rae, C. D. J. Proteome
(117) MacKinnon, N.; Ge, W.; Khan, A. P.; Somashekar, B. S.; Res. 2011, 10, 1737−1745.
Tripathi, P.; Siddiqui, J.; Wei, J. T.; Chinnaiyan, A. M.; Rajendiran, T. (147) Maher, A. D.; Fonville, J. M.; Coen, M.; Lindon, J. C.; Rae, C.
M.; Ramamoorthy, A. Anal. Chem. 2012, 84, 5372−5379. D.; Nicholson, J. K. Anal. Chem. 2012, 84, 1083−1091.
(118) Vu, T. N.; Valkenborg, D.; Smets, K.; Verwaest, K. A.; (148) Cloarec, O.; Dumas, M. E.; Craig, A.; Barton, R. H.; Trygg, J.;
Dommisse, R.; Lemiere, F.; Verschoren, A.; Goethals, B.; Laukens, K. Hudson, J.; Blancher, C.; Gauguier, D.; Lindon, J. C.; Holmes, E.;
BMC Bioinformatics 2011, 12, 405. Nicholson, J. Anal. Chem. 2005, 77, 1282−1289.
(119) Wang, B.; Goodpaster, A. M.; Kennedy, M. A. Chemom. Intell. (149) Posma, J. M.; Garcia-Perez, I.; De Iorio, M.; Lindon, J. C.;
Lab. Syst. 2013, 128, 9−16. Elliott, P.; Holmes, E.; Ebbels, T. M. D.; Nicholson, J. K. Anal. Chem.
(120) Fages, A.; Pontoizeau, C.; Jobard, E.; Levy, P.; Bartosch, B.; 2012, 84, 10694−10701.
Elena-Herrmann, B. Anal. Bioanal. Chem. 2013, 405, 8819−8827. (150) Barding, G. A., Jr.; Orr, D. J.; Sathnur, S. M.; Larive, C. K. Anal.
(121) Sousa, S. A. A.; Magalhaes, A.; Ferreira, M. M. C. Chemom. Bioanal. Chem. 2013, 405, 8409−8417.
Intell. Lab. Syst. 2013, 122, 93−102. (151) Wang, B.; Shi, Z. Q.; Weber, G. F.; Kennedy, M. A. Anal.
(122) Hao, J.; Liebeke, M.; Astle, W.; De Iorio, M.; Bundy, J. G.; Bioanal. Chem. 2013, 405, 8419−8429.
Ebbels, T. M. D. Nat. Protoc. 2014, 9, 1416−1427. (152) Shao, X. G.; Miao, L. N.; Liu, Z. C.; Liu, H. L.; Cai, W. S.
(123) Ye, T.; Zheng, C.; Zhang, S. C.; Gowda, G. A. N.; Vitek, O.; Spectrosc. Lett. 2011, 44, 244−250.
Raftery, D. Anal. Chem. 2012, 84, 994−1002. (153) Smolinska, A.; Blanchet, L.; Buydens, L. M. C.; Wijmenga, S. S.
(124) Yilmaz, A.; Nyberg, N. T.; Jaroszewski, J. W. Anal. Chem. 2011, Anal. Chim. Acta 2012, 750, 82−97.
83, 8278−8285.
(125) Wu, H. D. I.; Fushing, H. Stat. Interface 2011, 4, 443−450.
(126) Romano, R.; Canonico, R.; Acernese, F.; Indovina, P. L.;
Barone, F. Proc. SPIE, Health Monit. Struct. Biol. Syst. 2013, 8695,
869538.
(127) Mihaleva, V. V.; Korhonen, S. P.; van Duynhoven, J.; Niemitz,
M.; Vervoort, J.; Jacobs, D. M. Anal. Bioanal. Chem. 2014, 406, 3091−
3102.

146 dx.doi.org/10.1021/ac504075g | Anal. Chem. 2015, 87, 133−146

You might also like