2018 Book MetabolicProfiling
2018 Book MetabolicProfiling
2018 Book MetabolicProfiling
Georgios A. Theodoridis
Helen G. Gika
Ian D. Wilson Editors
Metabolic
Profiling
Methods and Protocols
Methods in Molecular Biology
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Georgios A. Theodoridis
Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
Helen G. Gika
School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
Ian D. Wilson
Department of Surgery and Cancer, Imperial College London, London, UK
Editors
Georgios A. Theodoridis Helen G. Gika
Department of Chemistry School of Medicine
Aristotle University of Thessaloniki Aristotle University of Thessaloniki
Thessaloniki, Greece Thessaloniki, Greece
Ian D. Wilson
Department of Surgery and Cancer
Imperial College London
London, UK
This Humana Press imprint is published by the registered company Springer Science+Business Media, LLC part of
Springer Nature.
The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A.
Preface
This book provides a number of protocols for “global metabolic profiling,” also known as
metabonomics and/or metabolomics. Metabolomics deals with the holistic analysis of small
molecules aiming to characterize the metabolic content of the studied samples/systems and
reveal changes that result from alterations to them as a result of, e.g., different physiological
states or the onset and progression of disease, etc. Over the last few decades, there have
been significant developments in both analytical technologies and multivariate statistical
methods that have greatly facilitated the growth of these holistic analytical approaches.
In putting together this volume, the editors have placed emphasis on obtaining chap-
ters that illustrate the different approaches taken by researchers to develop tools to address
the important challenges of the field. The first part of the book contains chapters on the
challenges and perspective of the topic (Gika et al.), the use of quality control measures
(QC) and validation issues (Begou et al.), data mining (Riccadonna and Francheschi), and
bio- and chemoinformatic tools for metabolomics (Witting). These chapters highlight basic
concepts such as experimental design, data treatment, metabolite identification, the need
for harmonization, and the linking of data obtained by different analytical modes (also
combining metabolomics results with data from other omics fields).
The second section, which is concerned with methodology, describes protocols for sam-
ple preparation centered on techniques for tissues, feces, and blood samples (Michopoulos,
Deda et al., also addressed by Vorkas et al.) and chemical derivatization for GC-MS (Hušek
et al.). The methods used for metabolite analysis and profiling are covered with chapters on
GC-MS metabolic profiling (Klapa et al.), LC-MS profiling using both targeted methods
(Virgiliou et al.) and IPC-LC-MS (Michopoulos), and untargeted (Want) profiling
approaches. The profiling of polar charged metabolites still remains a challenge, and this
section includes a chapter on the use of CE-MS for this purpose (Ramautar). NMR spectro-
scopic methods for profiling biological fluids (Benaki and Mikros) are also considered.
The volume concludes with two application sections covering the use of metabolomics
in life sciences with examples of methodologies that can be found in food science or bio-
marker discovery for disease diagnosis and human well-being. In the case of food and natu-
ral products, the protocols describe the analytical methods used and their application in
food quality control, where the use of NMR spectroscopy is described (Schripsema and
Dagnino) and the evaluation of product authenticity and geographical origin (Spyros and
coworkers). Both these issues represent major challenges for the food industry and are still
a great concern for the health of the consumer. The use of proton-transfer-reaction time-
of-flight mass spectrometry (PTR-TOF-MS) for the analysis of volatile organic chemicals
(VOCs) is also described (Farneti). Arapitsas and Mattivi describe a protocol on the analysis
of wine by LC-MS with application to the classification of wine according to the grape
variety. In the case of applications in life sciences, the use of metabolic profiling for bio-
marker discovery in cardiovascular disease (Vorkas et al.) and the targeted analysis of ste-
roids (Rudaz and coworkers) are described. Finally, Siopi and Mougios discuss experimental
design and considerations on sample collection for studies involving human subjects.
v
vi Preface
While still an area of rapid technical development, the place of “omic” metabolic phe-
notyping where the objective is to gain unbiased, global knowledge of the content of the
studied system, is firmly fixed as a means of gaining insights into the conditions under
study, thereby enhancing our knowledge and detailed understanding of the phenomena
under investigation.
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
Part I Fundamentals
Part II Methods
vii
viii Contents
ix
x Contributors
Fundamentals
Chapter 1
Abstract
Metabolic profiling has advanced greatly in the past decade and evolved from the status of a research topic
of a small number of highly specialized laboratories to the status of a major field applied by several hun-
dreds of laboratories, numerous national centers, and core facilities. The present chapter provides our view
on the status of the remaining challenges and a perspective of this fascinating research area.
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_1, © Springer Science+Business Media, LLC, part of Springer Nature 2018
3
4 Helen G. Gika et al.
Fig. 1 Publication trends in the areas of genomics/transcriptomics, proteomics, and metabolic phenotyping
from 2000 to 2016 generated from references contained within SCOPUS using the search terms genomics,
proteomics, metabolomics or metabonomics or metabolic profiling, and transcriptomics (search made June
2017)
Fig. 2 Representative 1H-NMR spectra of hepatic extract metabolic profiles of the acetaminophen (APAP),
AMAP, and control groups at 1 h. Resonances assigned to drug-related molecules have been colored in red.
Key: APAP/AMAP-G APAP/AMAP glucuronide, APAP-SG APAP glutathionyl, APAP-NAC APAP-N-acetylcysteinyl,
APAP/AMAPNHCOCH3 APAP/AMAP N-acetyl resonance, GSH reduced glutathione, GSSG oxidized glutathione,
Phe phenylalanine, d-3-HB d-3-hydroxybutyrate, AMP adenosine monophosphate, overlapped resonances
from glucose/glycogen/maltose labeled. From reference [12] reprinted with permission
Metabolic Profiling: Status, Challenges, and Perspective 7
solution being suitable for some, but not all, such compounds [19,
20]. For polar/ionic compounds that are unsuitable for analysis by
HILIC-MS, the only remaining option may be to use ion pair (IP)
LC where a suitable charged molecule, e.g., tributyl ammonium, is
added to the mobile phase as an oppositely charged counter ion to
“pair” with the oppositely charged analytes [21]. But, while effec-
tive, the use of IPLC generally requires the (effectively) permanent
dedication of the system to this mode of operation thereafter, as
decontaminating the instrumentation to remove all traces of the
IP-reagent can be challenging. An alternative to LC-based meth-
ods for polar ionic compounds is, of course, to employ a capillary
electrophoresis for the separation, and CE-MS methods have
indeed shown utility in this role [22].
The upshot of all of this is that, in order to obtain the most
comprehensive metabolite profile of a sample set possible, it may
require more than one chromatographic system and analysis in
both positive and negative modes of ionization (generally electro-
spray ionization (ESI), but also possibly APCI).
Having developed a suitable separation, a number of chal-
lenges remain in order to ensure that the analytical data that are
obtained are useful. Unlike NMR spectroscopy-based methods,
which are generally very robust, those utilizing a hyphenated MS
have a number of challenges that need to be addressed. These
result from the tendency of the analytical system (column and
detector) to become modified over the course of the analysis. This
can lead to minor changes in retention time, sensitivity, and (less
often) mass accuracy. The existence of such effects requires the use
of careful quality control procedures that can be used to monitor
the analysis and correct for analytical drift of whatever sort. Various
methods have been proposed for ensuring the validity of the data,
of which one of the most common is the use of so-called quality
control or QC samples. These are most often generated by making
a representative bulk pool sample from aliquots of the samples to
be analyzed. Typically it is first necessary to equilibrate the LC
system by the repeated injection of a number of QC samples, which
results in stable retention times. After this the QC sample is injected
at regular intervals throughout the sample analysis [23]. After the
run is completed, the data from these QC samples can be analyzed
using multivariate statistical procedures such as principal compo-
nent analysis (PCA), which provides a powerful tool to reveal
trends in data that would indicate time-related (or other) effects
that compromise the outcome. Assuming that the data passes such
preliminary scrutiny, further measures to optimize it, e.g., peak
alignment, can be performed, and the data can be examined for the
presence of potential biomarkers. This part of the process relies
heavily on the correct choice and the correct function of software
tools. Some software still operate as black boxes, not providing
10 Helen G. Gika et al.
References
1. Nicholson JK, Lindon JC, Holmes E (1999) and its less toxic meta-isomer. Arch Toxicol
“Metabonomics”: understanding the meta- 90:3073–3085. https://doi.org/10.1007/
bolic responses of living systems to patho- s00204-015-1655-x
physiological stimuli via multivariate statistical 13. Han J, Danell RM, Patel JR et al (2008)
analysis of biological NMR spectroscopic data. Towards high-throughput metabolomics using
Xenobiotica 29:1181–1189 ultrahigh-field Fourier transform ion cyclotron
2. Fiehn O, Kopka J, Dörmann P et al (2000) resonance mass spectrometry. Metabolomics
Metabolite profiling for plant functional 4:128–140
genomics. Nat Biotechnol 18:1157–1161 14. Theodoridis G, Gika HG, Wilson ID (2011)
3. Gavaghan CL, Holmes E, Lenz E et al (2000) Mass spectrometry-based holistic analytical
An NMR-based metabonomic approach to approaches for metabolite profiling in systems
investigate the biochemical consequences of biology studies. Mass Spectrom Rev 30:884–
genetic strain differences: application to the 906. https://doi.org/10.1002/mas.20306
C57BL10J and Alpk:ApfCD mouse. FEBS 15. Kopka J (2006) Current challenges and devel-
Lett 484:169–174 opments in GC–MS based metabolite profiling
4. Dent CE (1952) Lectures on the scientific basis technology. J Biotechnol 124:312–322
of medicine, vol 2. Athlone Press, London 16. Moros G, Chatziioannou AC, Gika HG et al
5. Dalgliesh CE (1956) Two-dimensional paper (2017) Investigation of the derivatization con-
chromatography of urinary indoles and related ditions for GC–MS metabolomics of biological
substances. Biochem J 64:481–485 samples. Bioanalysis 9:53–65
6. Teranishi R, Mon TR, Robinson AB et al 17. Gika HG, Theodoridis GA, Plumb RS et al
(1972) Gas chromatography of volatiles from (2014) Current practice of liquid chroma-
breath and urine. Anal Chem 44:18–20 tography–mass spectrometry in metabolomics
7. Pauling L, Robinson AB, Teranishi R et al and metabonomics. J Pharm Biomed Anal
(1971) Quantitative analysis of urine vapor and 87:12–25
breath by gas-liquid partition chromatography. 18. Rainville PD, Theodoridis G, Plumb RS et al
Proc Natl Acad Sci U S A 68:2374–2376 (2014) Advances in liquid chromatogra-
8. Robinson AB, Pauling L (1974) Techniques phy coupled to mass spectrometry for meta-
of orthomolecular diagnosis. Clin Chem bolic phenotyping. TrAC Trends Anal Chem
20:961–965 61:181–191. https://doi.org/10.1016/j.
9. Scott CD, Chilcote DD, Lee NE (1972) trac.2014.06.005
Coupled anion and cation-exchange chroma- 19. Theodoridis GA, Gika HG, Want EJ et al
tography of complex biochemical mixtures. (2012) Liquid chromatography–mass spec-
Anal Chem 44:85–89 trometry based global metabolite profiling: a
10. Scott CD, Chilcote DD, Katz S et al (1973) review. Anal Chim Acta 711:7–16
Advances in the application of high resolution 20. Gika HG, Wilson ID, Theodoridis GA (2014)
liquid chromatography to the separation of LC–MS-based holistic metabolic profiling.
complex biological mixtures. J Chromatogr Sci Problems, limitations, advantages, and future
11:96–100 perspectives. J Chromatogr B Analyt Technol
11. Lenz EM, Wilson ID (2007) Analytical strat- Biomed Life Sci 966:1–6
egies in metabonomics J. Proteome Res 21. Michopoulos F, Whalley N, Theodoridis G
6:443–458 et al (2014) Targeted profiling of polar intra-
12. Kyriakides M, Maitre L, Stamper BD et al cellular metabolites using ion-pair-high perfor-
(2016) Comparative metabonomic analysis mance liquid chromatography and-ultra high
of hepatotoxicity induced by acetaminophen performance liquid chromatography coupled
to tandem mass spectrometry: applications to
Metabolic Profiling: Status, Challenges, and Perspective 13
serum, urine and tissue extracts. J Chromatogr an HPLC−MS-based method for metabo-
A 1349:60–68 nomic analysis: application to human urine.
22. Ramautar R, Nevedomskaya E, Mayboroda J Proteome Res 6:3291–3303
OA et al (2011) Metabolic profiling of 24. h t t p s : / / e n . w i k i p e d i a . o r g / w i k i /
human urine by CE-MS using a positively Lies,_damned_lies,_and_statistics
charged capillary coating and comparison with 25. Xia J, Wishart DS (2010) MetPA: a web-based
UPLC-MS. Mol BioSyst 7:194–199. https:// metabolomics tool for pathway analysis and
doi.org/10.1039/c0mb00032a visualization. Bioinformatics 26:2342–2344.
23. Gika HG, Theodoridis GA, Wingate JE https://doi.org/10.1093/bioinformatics/
et al (2007) Within-day reproducibility of btq418
Chapter 2
Abstract
Global metabolic profiling (untargeted metabolomics) of different and complex biological matrices aims
to implement an holistic, hypothesis-free analysis of (potentially) all the metabolites present in the analyzed
sample. However, such an approach, although it has been the focus of great interest over the past few
years, still faces many limitations and challenges, particularly with regard to the validation and the quality
of the obtained results. The present protocol describes a quality control (QC) procedure for monitoring
the precision of the analytical process involving untargeted metabolic phenotyping of urine and plasma/
serum. The described/suggested methodology can be applied to different biological matrices, such as
biological biofluids, cell, and tissue extracts.
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_2, © Springer Science+Business Media, LLC, part of Springer Nature 2018
15
16 Olga Begou et al.
2 Materials
2.2 Mobile Phase –– Mobile Phase A: Water + 0.1 vol.% formic acid: add 1 mL of
formic acid to 1 L of Millipore water.
–– Mobile Phase B: Acetonitrile + 0.1 vol.% formic acid: add
100 μL of formic acid to 1 L of LC-MS grade acetonitrile. In
the case of serum samples use methanol instead of acetonitrile
or mixture of methanol with acetonitrile as mobile phase B.
–– Wash Solvent: Use a “strong” solvent acetonitrile/water
80:20 v/v for post-injection cleaning cycles and a weak solvent
water/acetonitrile 80:20 v/v for pre-injection washes (see
Note 2).
18 Olga Begou et al.
2.4 Software
Appropriate software include but are not limited to the following:
Data acquisition and processing software (e.g., Excalibur,
MassLYnx, Analyst, or other). Special software for peak picking
such as Marker Lynx, Sieve, MarkerView (or other vendor soft-
ware), or open-source/free software (XCMS, MzMine, MetAlign
or other). Microsoft Excel, Statistica, and other advanced spread-
sheet software. SIMCA-P or other software for multivariate statis-
tical analysis. MATLAB software and programming language or
associated software packages: The programming language R is a
popular and easy solution for data analysis, statistical computing,
and graphics support.
3 Methods
3.1 Analytical 1. Before starting, ensure that the mass spectrometer is in a suit-
System Preparation able condition for the analysis of the samples. Check the mass
accuracy of the mass spectrometer; if necessary, calibrate the
mass spectrometer following the appropriate procedure recom-
mended by the vendor to achieve maximum mass accuracy and
resolution. It is advisable that the calibration procedure should
take place every 3–4 months and that the temperature of the
laboratory should not vary significantly.
2. Load the required liquid chromatographic method by setting
parameters such as flow rate, column and autosampler tempera-
ture, wash cycles, and gradient elution program.
3. Allow the system to equilibrate and run a no-injection gradient
in order to aid column equilibration. Carefully examine the
results for this blank run for evidence of system/solvent con-
tamination etc., and decontaminate if necessary.
4. Depending on the matrix of the samples being analyzed, run a
suitable number of QC samples (prepared as described below)
to achieve system stability. It has been observed that for urine
analysis, 5 QC replicates are needed and for serum or plasma
10–20 injections (see Notes 3 and 4).
Quality Control and Validation Issues in LC-MS Metabolomics 19
3.2 Sample Handling 1. It is advisable to always divide all samples on collection into an
appropriate number of sub-aliquots for later storage and also in
order to avoid unnecessary freeze/thaw cycles.
2. If possible, store sub-aliquots in different freezers, in case a
freezer malfunction takes place. Ideally, samples should be
stored in freezers as soon as possible after sampling/collection
and at the lowest available temperature, at least at −20 °C but
preferably at −80 °C.
3. Make sure all samples, stock, and working solutions are cor-
rectly and fully labeled. Thaw stock/working solutions and real
samples shortly before use and sample preparation,
respectively.
3.3 Sample 1. Vigorously vortex every sample, after thawing at room tem-
Preparation perature, prior to sample preparation.
2. Transfer an appropriate volume (e.g., 50–200 μL) of each sam-
ple into 1.5 mL Eppendorf tubes.
3. Depending on the matrix (urine, plasma, serum), a different
sample preparation procedure is recommended (see Notes 5
and 6) as explained below in Subheadings 3.3.1–3.3.3.
3.3.1 For Urine Samples
1. Dilute the sample in ratio 1:3 v/v with Millipore quality water.
2. Vortex for 5 min and centrifuge at 12,000 × g or higher speeds
for 5–10 min at 4 °C.
3. Transfer the clear supernatant into an LC-MS vial (n.b. 96 well
plates or similar can be substituted for LC-MS vials).
4. Place the vial (well plate) into a pre-cooled (4 °C) autosampler
for analysis. If analysis is not being undertaken immediately,
store the vial (well plate) in a fridge (0–4 °C) if analysis will start
shortly or a freezer (−20/80 °C) if analysis will be delayed for
longer than a few hours.
3.3.2 For Plasma or
Serum Samples 1. Add three times the sample volume of ice cold methanol or
acetonitrile for protein precipitation (see Note 7).
2. Vortex for 5 min and centrifuge at 12,000 × g or higher speeds
for 5–10 min.
3. Transfer the clear supernatant into Eppendorf tubes.
4. Evaporate to dryness and reconstitute with water/acetonitrile
95:5 v/v to the initial volume.
5. Repeat step 2.
6. Transfer the clear supernatant into an LC-MS vial (well plate).
7. Place the vial (well plate) into a pre-cooled (4 °C) autosampler
for analysis. If analysis is not taking place immediately, follow
20 Olga Begou et al.
3.4 Analytical 1. Make sure that the order of the samples is randomized, to avoid
Sequence Preparation introducing bias due to changes with individual analytical runs
and between run “batch” effects. Randomization of large sam-
ple sets can be performed using the specific commands in
spreadsheet programs such as Excel. QC samples should not be
randomized but inserted regularly in the run sequence (e.g.,
one QC sample every five to ten real samples).
2. The number of QC samples placed in a sequence depends on
the total number of samples analyzed and on the duration of
the analysis. For a small number of samples (<100), one QC
every five sample injections is recommended, and for a larger
number QC, samples should represent at least 10% of the total
analyzed.
3. After column equilibration, in the middle of the sequence and
at the end, insert the spiked test mix solution. Standard solution
injections should be avoided within the batch of test samples, as
the system can be disequilibrated by the injection of non-matrix
Quality Control and Validation Issues in LC-MS Metabolomics 21
Thereinafter, in the exported peak table data, look for any sim-
ilarities in the pairs (retention time and mass) among the peaks
reported, in order to assess the success of peak alignment. Then,
using appropriate statistical software, create graphs with the data
exported from the QC samples to evaluate the analytical perfor-
mance and precision.
Figure 1 presents a schematic of the proposed validation
scheme. It starts with the QC preparation, by mixing equal vol-
umes of all the real samples (urine, plasma/serum, etc.) followed
by determing the run order sequence and then injecting the QC
sample periodically, e.g., every five to ten real samples during anal-
ysis. From the resulting data set, an extracted ion chromatogram
(XIC) of all QC samples can be created, via data processing or data
mining, where data is collected only for specific m/z values of the
compounds of interest. Those results can also be used for the for-
mation of an analysis chart, showing total sequence order (both
QC and real samples). Data processing is complemented by statis-
tical analysis. Repeatability and run order trends can be assessed by
making QC quality control charts and data tables, evaluating %RSD
values of QC’s ion intensities. Features presented in most samples
(~70%) with %RSD values <30% indicate a good data set. Finally,
PCA score plots can be created from the examined results, provid-
ing information on the accuracy and precision of the analysis, as
well as for any time trends related to the order of analysis of both
QC and test samples.
4 Notes
Fig. 1 Schematic of a validation scheme from QC and sequence preparation (top), to data processing (middle)
to statistical analysis (bottom). From top left: QC samples are prepared by initially mixing equal volume of all
the real samples. Data processing includes scrutiny of data as extracted ion chromatogram (XIC) and data
processing or data mining. Trends in data should be checked across the run order. Repeatability and run order
trends can be assessed by making QC quality control charts and data tables, evaluating %RSD values of QC’s
ion intensities. PCA score plots should exhibit tight cloud of QC samples (red in the presented plot) within the
whole sample set (black dots). In the bottom of the figure, a control chart (left) shows features showing poor
repeatability in red (CV >30% in QC samples) and repeatable features in green (CV <30% in QC samples).
These charts are providing information for the precision of the analysis, as well as for any time trends related
to the order analysis of both QC and real samples (bottom right)
References
1. Nicholson JK, Lindon JC, Holmes E (1999) of day-to-day reproducibility and robustness of
“Metabonomics”: understanding the meta- LC-MS methods for global metabolite profiling
bolic responses of living systems to patho- in metabonomics/metabolomics. Bioanalysis
physiological stimuli via multivariate statistical 4:2239–2247. https://doi.org/10.4155/
analysis of biological NMR spectroscopic data. bio.12.212
Xenobiotica 29:1181–1189. https://doi. 9. Begou O, Gika HG, Wilson ID et al (2017)
org/10.1080/004982599238047 Hyphenated MS-Based Targeted approaches in
2. Dunn WB, Wilson ID, Nicholls AW, metabolomics. Analyst 142(17):3079–3100
Broadhurst D (2012) The importance of 10. Theodoridis G, Gika HG, Wilson ID (2011)
experimental design and QC samples in large- Mass spectrometry-based holistic analytical
scale and MS-driven untargeted metabolomic approaches for metabolite profiling in sys-
studies of humans. Bioanalysis 4:2249–2264. tems biology studies. Mass Spectrom Rev
https://doi.org/10.4155/bio.12.204 30:884–906. https://doi.org/10.1002/
3. Griffiths WJ, Koal T, Wang Y et al (2010) mas.20306
Targeted metabolomics for biomarker dis- 11. Gika HG, Zisi C, Theodoridis G et al (2016)
covery. Angew Chem Int Ed 49:5426–5445. Protocol for quality control in metabolic pro-
https://doi.org/10.1002/anie.200905579 filing of biological fluids by U(H)PLC-MS. J
4. Wilson I (2016) Methods and techniques for Chromatogr B Analyt Technol Biomed Life
metabolic phenotyping. Bioanalysis 9:1–3. Sci 1008:15–25. https://doi.org/10.1016/j.
https://doi.org/10.4155/bio-2016-4985 jchromb.2015.10.045
5. Gika HG, Wilson ID, Theodoridis GA (2014) 12. Gorrochategui E, Jaumot J, Lacorte S et al
LC–MS-based holistic metabolic profiling. (2016) Data analysis strategies for targeted and
Problems, limitations, advantages, and future untargeted LC-MS metabolomic studies: over-
perspectives. J Chromatogr B 966:1–6. https:// view and workflow. TrAC Trends Anal Chem
doi.org/10.1016/j.jchromb.2014.01.054 82:425–442. https://doi.org/10.1016/j.
6. Gika HG, Theodoridis GA, Earll M et al trac.2016.07.004
(2010) Does the mass spectrometer define 13. Naz S, Vallejo M, García A et al (2014)
the marker? A comparison of global metabo- Method validation strategies involved in non-
lite profiling data generated simultaneously via targeted metabolomics. J Chromatogr A
UPLC-MS on two different mass spectrom- 1353:99–105. https://doi.org/10.1016/j.
eters. Anal Chem 82:8226–8234. https://doi. chroma.2014.04.071
org/10.1021/ac1016612 14. Bijlsma S, Bobeldijk I, Verheij ER et al (2006)
7. Rainville PD, Theodoridis G, Plumb RS, Wilson Large-scale human metabolomics studies: a
ID (2014) Advances in liquid chromatogra- strategy for data (pre-) processing and valida-
phy coupled to mass spectrometry for meta- tion. Anal Chem 78:567–574. https://doi.
bolic phenotyping. TrAC Trends Anal Chem org/10.1021/ac051495j
61:181–191. https://doi.org/10.1016/j. 15. Want EJ, Wilson ID, Gika H et al (2010)
trac.2014.06.005 Global metabolic profiling procedures for urine
8. Gika HG, Theodoridis GA, Earll M, Wilson ID using UPLC-MS. Nat Protoc 5:1005–1018.
(2012) A QC approach to the determination https://doi.org/10.1038/nprot.2010.50
26 Olga Begou et al.
16. Gika HG, Theodoridis GA, Wingate JE for precision large scale urinary metabolic phe-
et al (2007) Within-day reproducibility of notyping. Anal Chem 88:9004–9013. https://
an HPLC-MS-based method for metabo- doi.org/10.1021/acs.analchem.6b01481
nomic analysis: application to human urine. 20. Veselkov KA, Vingara LK, Masson P et al
J Proteome Res 6:3291–3303. https://doi. (2011) Optimized preprocessing of ultra-
org/10.1021/pr070183p performance liquid chromatography/mass
17. Zelena E, Dunn WB, Broadhurst D et al spectrometry urinary metabolic profiles for
(2009) Development of a robust and repeatable improved information recovery. Anal Chem
UPLC−MS method for the long-term metab- 83:5864–5872. https://doi.org/10.1021/
olomic study of human serum. Anal Chem ac201065j
81:1357–1364. https://doi.org/10.1021/ 21. Michopoulos F, Theodoridis G, Smith CJ,
ac8019366 Wilson ID (2010) Metabolite profiles from
18. Dunn WB, Broadhurst D, Begley P, Human dried biofluid spots for metabonomic stud-
Serum Metabolome (HUSERMET) Consortium ies using UPLC combined with oaToF-MS. J
(2011) Procedures for large-scale metabolic pro- Proteome Res 9:3328–3334. https://doi.
filing of serum and plasma using gas chromatog- org/10.1021/pr100124b
raphy and liquid chromatography coupled to mass 22. Ramautar R, Nevedomskaya E, Mayboroda
spectrometry. Nat Protoc 6:1060–1083. https:// OA et al (2011) Metabolic profiling of
doi.org/10.1038/nprot.2011.335 human urine by CE-MS using a positively
19. Lewis MR, Pearce JTM, Spagou K et al charged capillary coating and comparison with
(2016) Development and application of ultra- UPLC-MS. Mol BioSyst 7:194–199. https://
performance liquid chromatography-TOF MS doi.org/10.1039/c0mb00032a
Chapter 3
Abstract
Liquid chromatography-mass spectrometry (LC-MS) untargeted experiments require complex chemo-
metrics strategies to extract information from the experimental data. Here we discuss “data preprocess-
ing”, the set of procedures performed on the raw data to produce a data matrix which will be the starting
point for the subsequent statistical analysis. Data preprocessing is a crucial step on the path to knowledge
extraction, which should be carefully controlled and optimized in order to maximize the output of any
untargeted metabolomics investigation.
Key words Preprocessing, Peak picking, Retention time correction, Metadata, Quality check, Missing
values
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_3, © Springer Science+Business Media, LLC, part of Springer Nature 2018
27
28 Samantha Riccadonna and Pietro Franceschi
Question Preprocessing
Definition
Data conversion
Data generation Peak picking
(open file format)
Design of
Grouping and retention time correction
Experiment
(DoE)
Metadata
Quality check
organization
Explorative Data
Analysis transformation
Result Validation
& Interpretation
Fig. 1 Data analysis workflow of a typical LC-MS untargeted experiment: the path from data to knowledge is
usually represented as linear, but it is more likely to have many feedback loops (dashed lines). Only the red
boxes are detailed in the text
2 Software
3 Methods
3.1 Metadata The term “experiment metadata” refers to the set of information
Organization describing the samples and the analytical pipeline (see Note 2).
Organizing and storing a detailed record of the sample metadata
(see Note 3) is crucial to perform a reliable interpretation of the
results, evaluating the presence (and the impact) of possible con-
founding factors.
1. Collect and organize (possibly with the help of a spreadsheet)
all the important properties of the samples (e.g. treatment type,
day of collection, origin, etc.). Keep track also of the sample
specific analytical details.
2. Run ISAcreator to generate your ISA-Tab files (see Note 4).
The tool will support the description, through commonly
accepted “ontologies”, of the general aim of your experiment
(“Study”), the analytical platform (“Study Assay”), the DoE
(“Study Design Descriptors”, “Study Factors”), the analytical
information (“Study Protocols”) and the details about each
sample (“Sample Definitions”). Handling large datasets with
ISAcreator may not be easy, in particular if one wants to auto-
matically fill some of the required fields. In this case, an alterna-
tive choice is to use ISAcreator only to generate the backbone
of the ISA-Tab files and to fill in the sample details either by a
spreadsheet or a scripting language (e.g. R, python, etc.). In R,
30 Samantha Riccadonna and Pietro Franceschi
3.2 Data Conversion In general, the machine software can be used to perform this task
(See Note 6) (see Note 7). At present, the open source alternative is the
ProteoWizard “msconvert” (see Note 8), which is available as a
command line tool or as a point-and-click application for Windows
(MSconvertGUI) (see Note 9).
1. Open a terminal or a command window and run msconvert
into the raw data directory. As an example, to convert vendor-
specific raw data (here .RAW data files) into mzML files and
store them into the “open_data” directory use:
msconvert *.RAW -o open_data.
Alternatively, the use of the GUI application is self-explanatory.
Also in this case, batch mode conversion of a set of files is
allowed.
3.3 Processing Each metabolite produces at least one peak in the (m/z, rt) plane. To
detect metabolites, an automatic algorithm has to be able to identify
such peaks and to distinguish them from electrical or chemical noise
[7, 21]. This process of peak identification (peak picking) has to be
performed automatically on all the samples under analysis.
Subsequently, the peak lists identified in each sample have to be
joined together in a final list of features. To do that, it is necessary to
account for m/z and rt shifts from sample to sample [22]. The mea-
sured m/z values can be different because even the most advanced
mass spectrometer is not absolutely precise. Rt shifts, instead, are
unavoidable characteristics of chromatography. Technically speak-
ing, a “retention time correction” step is implemented to compen-
sate for the shifts in rt, and, afterwards, the features found in the
different samples are “grouped” (across samples) in a final consensus
list. All the software solutions (commercial or open source) imple-
ment different strategies to perform these steps [23, 24].
Data Treatment for LC-MS Untargeted Analysis 31
3.4 Preprocessing: The characteristics that distinguish a peak from the noise are inten-
Peak Picking sity, width and shape. Different algorithms handle them in differ-
ent ways [7, 21]. Most importantly, not only the algorithm but
also the specific parameters of each algorithm greatly affect the
results (see Note 10).
1. Choose the peak-picking algorithm on the basis of the charac-
teristics of the analytical pipeline [25, 26]. In xcms several
choices are already available (see Note 11). The type of instru-
mental setting used to analyse MTBLS59 suggests using the
centWave algorithm [18].
2. Identify the critical parameters of the algorithm and possibly
link them with the characteristics of the analytical pipeline. For
centWave, for example, the most important parameters to be
considered are ppm, which is determined by the mass accuracy
and is used to determine the “reliability” of each mass trace;
prefilter, which has to be set considering the minimum intensity
(I) of a true signal (at the single spectrum level) and also in how
many consecutive scans (k) that signal should be detected;
peakwidth, which deals with the range of the acceptable chro-
matographic peak-widths (in seconds); and snthresh, which
defines a cut-off for distinguishing signal from noise (signal-to-
noise ratio).
3. Check on a representative sample the results of the peak picking
(see Note 12). For instance, to run centWave with default
parameters on a specific file, issue
test_sample <- xcmsSet(files="MTBLS59/apple_
control_neg_001.CDF", method="centWave")
To visually inspect the outcomes of this step, plotting the peaks
in the m/z, rt plane, issue
mypeaks<-peaks(test_sample)
plot(mypeaks[,c("rt","mz")])
4. Optimize the peak-picking parameters until the result is satis-
factory (see Note 13).
5. Perform the peak picking on the whole dataset with the selected
set of parameters. The final xcmsSet object (see Note 11) will
contain the full list of the peaks detected in all the samples, with
some additional information on the mass and retention time
ranges. The complete analysis (with the parameters set as sug-
gested in [26]) can be run issuing:
cdffiles <- list.files(path="MTBLS59",pattern=
”CDF”,
full.names = TRUE)
myfiles<-grep("neg",cdffiles, value=TRUE)[1:40]
xs<-xcmsSet(files=myfiles,method="centWave",
ppm = 15, prefilter = c(0,0), peakwidth =
c(5,20), snthresh=1)
32 Samantha Riccadonna and Pietro Franceschi
3.5 Preprocessing: 1. Choose the retention time correction algorithm among the
Retention Time available options (see Note 11). In this example we use obiwarp
Correction [26], which is based on dynamic time warping, an algorithm
and Grouping (See used to find the best stretching of the time dimension of two
Note 14) time series to make them as similar as possible [27]. LC-MS
data are multidimensional, so in obiwarp the similarity is deter-
mined taking into account the full spectral information.
2. Determine the more relevant parameters of the algorithm. In
obiwarp the relevant parameters are the choice of the reference
sample for the rt warping (center); the m/z bins used for
retrieving the spectra (profStep); the spectral similarity function
(distFunc); and the penalizations on the warping optimization
(gapInit and gapExtend). With the default parameters (which
work reasonably well in most of the cases), the reference sample
is the one containing the largest number of peaks. The reten-
tion time correction can be performed with the following
command.
xsr <− retcor(xs, method=“obiwarp”)
3. Choose the “grouping” algorithm or, in other terms, the pro-
cedure which matches peaks with similar m/z and rt found in
different samples. Among the possible choices, we rely on the
default density-based solution [17].
4. Choose the grouping parameters according to the analytical
pipeline: bw defines the retention time window used for the
density estimation and mzwid accounts for small differences in
the m/z value of the corresponding peaks detected in the differ-
ent samples. To avoid the inclusion in the final list of features
peaks detected only in a small number of samples (which could
be, e.g. artefacts), it is possible to keep only groups which con-
tain peaks detected in at least minsamp samples (in at least one
sample class). Again, we set the parameters as suggested in [26]
and minsamp equal to 2:
xsr <− group(xsr, bw = 2, mzwid = 0.025,
minsamp = 2)
Retention Time
Fig. 2 Outcome of the retention time correction performed on the MTBLS59 data as obtained by plotrt, after
minor visual parameters customization. Each line represents one sample and is coloured by sample class. The
retention time correction is different for each sample, and it is not linear in time
1500
1000
Intensity
500
0
Fig. 3 Quality check on the processing procedure using the quercetin-3-glucoside and quercetin-3-galactoside
in negative ionization, which are expected to be present. Minor visual parameters customization has been
performed compared to the base command reported in the text. Samples are coloured by sample class. The
black dashed line corresponds to the identified the peak group
mygroups<−groups(xsr)
mycompound<−mygroups[mygroups[,"rtmed”]>405
& mygroups[,"rtmed”]<430
& mygroups[,"mzmed”]>463.082
& mygroups[,"mzmed”]<463.092,"rtmed”]
abline(v=mycompound)
The resulting plot is shown in Fig. 3. It is clear that the EICs are
correctly aligned (see Note 15). In this case, however, xcms
finds only one feature associated with both compounds
because of the initial choices of the peak-picking algorithm
(see Note 16).
3.7 Preprocessing: It could happen that peaks belonging to a subset of samples are
Imputation of Missing missing from “their” groups. This is correct if the correspond-
Peaks ing analyte is not present in a specific sample, but it could also
be the result of an error in the peak-picking phase. These miss-
ing peaks produce missing values in the data matrix, which are
usually tricky to handle during statistical analysis. Several
options exist to “impute” missing values (which are common
also in other -omic technologies). The clever solution available
in xcms is implemented by the function fillPeaks, which goes
back to the raw data integrating the signal measured in the area
corresponding to the “missing” peak. If the peak was wrongly
missed, fillPeaks will recover it, otherwise that missing value
will be filled with the sum of the noise.
Data Treatment for LC-MS Untargeted Analysis 35
3.8 Data Matrix Extract the final two-dimensional data matrix with samples on the
Extraction rows and features on the columns, using as intensity the integral of
the area under the peak:
mat<-t(groupval(xsf,value=“into”))
The m/z and rt details about the features are encoded in the
mygroups object generated at Subheading 3.6, step 2.
4 Notes
MTBLS59
0 200 400 600
default set1
2500
2000
1500
m/z
1000
500
rt
Fig. 4 Peak-picking outcome on the MTBLS59 dataset: (left) the peak detection is performed using the default
settings for centWave (right) or the set of parameters defined in Methods Subheading 3.4, step 5
References
21. Zhang J, Gonzalez E, Hestilow T et al (2009) 30. Bouyssié D, Dubois M, Nasso S et al (2015)
Review of peak detection algorithms in liquid- mzDB: a file format using multiple indexing
chromatography- mass spectrometry. Curr strategies for the efficient analysis of large
Genomics 10:388–401 LC-MS/MS and SWATH-MS data sets. Mol
22. Aberg KM, Alm E, Torgrip RJO (2009) The Cell Proteomics 14:771–781
correspondence problem for metabonomics 31. Krzywinski M, Altman N (2014) Points of sig-
datasets. Anal Bioanal Chem 394:151–162 nificance: designing comparative experiments.
23. Smith R, Ventura D, Prince JT (2015) LC-MS Nat Methods 11:597–598
alignment in theory and practice: a compre- 32. Krzywinski M, Altman N (2014) Points of sig-
hensive algorithmic review. Brief Bioinform nificance: analysis of variance and blocking. Nat
16:104–117 Methods 11:699–700
24. Koch S, Bueschl C, Doppler M et al (2016) 33. Krzywinski M, Altman N, Blainey P (2014)
MetMatch: a semi-automated software tool for the Points of significance: nested designs. Nat
comparison and alignment of LC-HRMS data Methods 11:977–978
from different metabolomics experiments. Meta. 34. Krzywinski M, Altman N (2014) Points of sig-
https://doi.org/10.3390/metabo6040039 nificance: two-factor designs. Nat Methods
25. Brodsky L, Moussaieff A, Shahaf N et al (2010) 11:1187–1188
Evaluation of peak picking quality in LC-MS 35. Altman N, Krzywinski M (2014) Points of sig-
metabolomics data. Anal Chem nificance: sources of variation. Nat Methods
82:9177–9187 12:5–6
26. Patti GJ, Tautenhahn R, Siuzdak G (2012) 36. Blainey P, Krzywinski M, Altman N (2014)
Meta-analysis of untargeted metabolomic data Points of significance: replication. Nat Methods
from multiple profiling experiments. Nat 11:879–880
Protoc 7:508–516 37. Altman N, Krzywinski M (2015) Points of sig-
27. Prince JT, Marcotte EM (2006) nificance: split plot design. Nat Methods
Chromatographic alignment of ESI-LC-MS 12:165–166
proteomics data sets by ordered bijective inter- 38. Haug K, Salek RM, Conesa P et al (2013)
polated warping. Anal Chem 78:6140–6152 MetaboLights–an open-access general-purpose
28. Martens L, Chambers M, Sturm M et al (2011) repository for metabolomics studies and associ-
mzML–a community standard for mass spec- ated meta-data. Nucleic Acids Res
trometry data. Mol Cell Proteomics 41:D781–D786
10:R110.000133 39. Libiseller G, Dvorzak M, Kleb U et al (2015)
29. Wilhelm M, Kirchner M, Steen JAJ, Steen H IPO: a tool for automated optimization of
(2012) mz5: space- and time-efficient storage XCMS parameters. BMC Bioinformatics
of mass spectrometry data sets. Mol Cell 16:118
Proteomics 11:O111.011379
Chapter 4
Abstract
Metabolomics data analysis includes several repetitive tasks, including data sorting, calculation of exact
masses or other physicochemical properties, or searching for identifiers in different databases. Several of
these tasks can be automated using command line tools or short scripts in different scripting languages like
Perl, Python, or R. This chapter presents simple solutions and short scripts written in R that can be used
for the interaction with specific web services or for the calculation of physicochemical properties or molec-
ular formulae.
Key words R, isotope pattern, Formula calculation, Physicochemical properties, Command line, Web
service, Identifier conversion
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_4, © Springer Science+Business Media, LLC, part of Springer Nature 2018
41
42 Michael Witting
in cells. In KEGG, acetate and acetic acid are stored under the
same KEGG id but have different accession numbers in
ChEBI. Cross mapping of different database identifiers is therefore
important and has to be updated on a regular basis and manually
annotated and corrected. The chemical translation service is an
open tool available to the metabolomics community, which offers
highly curated mappings between identifiers from databases that
are important for metabolomics [13].
This chapter presents approaches for the calculation of physi-
cochemical parameters, molecular formulae from exact masses,
generation of molecular structures, mapping of different database
identifiers and their conversion, as well as mapping of metabolites
onto pathways. Much emphasis is laid on (semi)automatic process-
ing using command line tools and short R scripts, based on previ-
ously published packages. Simple examples show the possibilities
and are also easy to use even for beginners.
2 Materials
2.1 Usage This chapter uses two different types of command line tools. Usage
of Command Line requires a few simple commands to handle the command line. The
Tools command line shell can be called in windows by using the windows
key and typing cmd and pressing the enter button. This should
open a command line in windows similar to the one shown in
Fig. 1. This chapter will solely focus on the Windows version of
commands.
First, if installed correctly, the ChemAxon tools should be
available as environmental variables and can be called by simply
typing cxcalc or molconvert. This can be tested by simply typing
cxcalc. If the following message appears, the environmental vari-
ables are not correctly set: “cxcalc is not recognized as an internal
or external command, operable program or batch file.” If this is
the case, please check the ChemAxon online manual on how to
integrate manually the environmental variables. The tools can be
also called directly from their installation folder, usually C:\Program
Files\ChemAxon\JChemSuite\bin.
The second possibility used in this chapter is to call java pack-
ages in a .jar file to perform calculations. For this, Java has to be
installed correctly and integrated in the environmental variables. .jar
44 Michael Witting
Fig. 1 (a) Example for the use of command line tools on a Windows system. The first lines demonstrate the use
of the cxcalc command from the ChemAxon JChem Suite for calculation of an exact mass, molecular formula,
and logP. The following lines show how to call the Parallel Molecule Generator java file to calculate all possible
structures for the molecular formula of leucine. (b) Screenshot of RStudio showing all basic windows. In the
scripting area, all commands can be collected in order to allow repetitive analysis without the need to recall all
commands. The global environment contains all currently active variables and allows their inspection for type
and size, while the output console allows to run single commands and prints all outputs
files are executed by calling it with the command java –jar XXX.jar
args, whereby XXX.jar refers to the .jar file to be used and args is
replaced by the argument that shall be passed to the program. An
example is shown in the lower part of Fig. 1. An important command
to remember is how to change between directories; this is done by
using the cd command, e.g., cd <pathToDirectory> changes into
the respective directory, while cd .. changes to next higher/lower
level in the directories, e.g., from C:\Users\TestUser\ to C:\Users.
Bio- and Chemoinformatics Approaches for Metabolomics Data Analysis 45
2.2 R Scripts All scripts can be used directly in R or more comfortably with an
IDE like RStudio, which allows more convenient work without the
need to remember all variables, since they are displayed in a sepa-
rate window and also allow manual inspection (Fig. 1b). The pre-
sented code snippets work with different packages which are not
part of the R base distribution and have to be therefore installed
manually. Specific packages are jsonlite, XML, rcdk, RCurl, curl,
rcdk, OrgMassSpecR, and CHNOSZ with their dependencies.
The following script will install all needed packages from CRAN
()https://cran.r-project.org/.
#list of packages needed
packages <- c("jsonlite", "XML", "rcdk",
"RCurl", "curl", "OrgMassSpecR", "CHNOSZ")
install.packages(packages, dependencies = T)
For the work with R scripts, a basic understanding of R func-
tionalities and data structures is needed (see Note 1). Most results
in the example scripts are returned as a list of specific datatypes,
e.g., formula generation with rcdk returns a list of the formal class
cdkFormula. Positions in this list are accessed via [i] or [[i]], where
i is the index of interest and specific information like the string
containing the molecular formula are stored in slots and can be
accessed via the @ operator.
#get formula string
formulae[[1]]@string
Another example is the use of JavaScript Object Notation,
short JSON, for transfer of structured data transfer, e.g., from web
services. This data format is used by many REST web services for
result representation. One example is the Chemical Translation
Service offered by the Fiehn Lab. A specific JSON result could
look like this.
[
{
"fromIdentifier": "inchikey",
"searchTerm": "QNAYBMKLOCPYGJ- REOHCLBHSA- N",
"toIdentifier": "Chemical Name",
"result":
[
"L-Alanine",
"L-2-Aminopropanoic acid"
]
}
]
The package jsonlite or the content() function from the httr
package allows a direct conversion of the results in a JSON string
to specific datatypes for a more convenient access, which is
described in a later section of this chapter.
46 Michael Witting
3 Methods
3.2 Calculation For people who would not like to use ChemAxon or are not
of Exact Mass, logD, allowed to use it, the Chemistry Development Kit (CDK) is a rich
and Others Using open-source alternative [14, 15]. However, it requires knowledge
RCDK on programming in Java, and no command line tools are available.
The package rcdk provides all CDK functionalities in R and can be
accessed easily. An important point is the correct installation of the
rJava package which is used to interface java functionalities with R
(see Notes 4 and 5).
1. First the required library is loaded.
#load required libraries
library(rcdk)
2. Next, a molecule to work with is generated by parsing a SMILES
string, in this case caffeine.
#parse smiles
smile <- 'Cn1cnc2c1c(=O)n(c(=O)n2C)C'
mol <- parse.smiles(smile)[[1]]
3. Before the molecule can be passed to the functions for the cal-
culation of exact mass and logP, it has to be prepared. This
includes detection of aromaticity, type of atoms, and configura-
tion of isotopes.
#prepare molecule
do.aromaticity(mol)
do.typing(mol)
do.isotopes(mol)
4. After the molecule has been prepared, the get.exact.mass(), get.
alogp(), get.xlogp(), get.tpsa(), and get.volume() functions can
be used for calculation of exact mass, logP, total polar surface
area, and the molecular volume.
#do the calculation
get.exact.mass(mol)
get.alogp(mol)
get.xlogp(mol)
get.tpsa(mol)
get.volume(mol)
Bio- and Chemoinformatics Approaches for Metabolomics Data Analysis 49
3.3 Formula rcdk offers possibilities useful for mass spectrometry-based metab-
and Isotope Pattern olomics, e.g., calculation of molecular formulas from masses, vali-
Calculation Using dation of molecular formulae, or calculation of isotope patterns. A
RCDK usual first step in metabolite identification is the calculation of
molecular formula from a measured exact mass and the compari-
son of theoretical and measured isotope patterns. Different soft-
ware tools exist for this purpose. The formula calculation and
validation is based on the seven golden rules proposed by Kind and
Fiehn [5].
50 Michael Witting
The following examples show how rcdk can be used for the
calculation of molecular formulae from a given exact mass.
1. First, the required library is loaded. In this first part, only rcdk
is required; however, the packages OrgMassSpecR and
CHNOSZ provide useful functionalities used later.
#load required libraries
library(rcdk)
library(OrgMassSpecR)
library(CHNOSZ)
2. The R script starts from a given isotopic pattern measured
([Glucose + Na]+). First, the monoisotopic mass is located (in
this case lowest m/z value) and is used as input for the generate.
formula() function of the rcdk package. This function has sev-
eral important parameters to set; first the search window has to
be set correctly; otherwise, the search for potential sum formu-
las is too exhaustive. Second, as stated in the seven golden rules,
the number of elements has to be restricted. Since the exact
mass used as input corresponds to a [M+Na] + adduct, sodium
has to be included; additionally, the parameter charge has to set
correct, in this case to +1.
#get measured isotope pattern ([Glucose +
Na]+)
isoPatternMeasured <- data.frame(mz =
c(203.052609, 204.056051, 205.057227,
206.060394, 207.061845),
int = c(100.000, 6.856, 1.433, 0.087,
0.009))
#get monoisotopic mass
exactmass <- isoPatternMeasured$mz[1]
#calculate all possible formulae
formulae <- generate.formula(exactmass, win-
dow = 0.001,
elements = list(c("C",0,10),c("H",0,50),
c("N",0,5),c("O",0,50),
c("Na",0,1)),
validation = T,
charge = 1)
3. In the first case, only one valid chemical formula is produced
and returned as a list of cdkFormula objects. Different values
can be accessed via the @ operator.
#get formula string
formulae[[1]]@string
#get charge
formulae[[1]]@charge
#get mass
formulae[[1]]@mass
Bio- and Chemoinformatics Approaches for Metabolomics Data Analysis 51
Fig. 2 (a) Enumeration of chemical structures using from structures generated by PMG containing a
cxcalc from the JChem tools allows to remove chemi- valence 5 nitrogen were removed, reducing possible
cal invalid structures, e.g., containing nitrogen atoms chemical structures from 32,395 to 24,843
of valence 5. In this particular example, all results candidates
3.5 Identifier The most unique identifier of a molecule is its structure. However,
Conversion using it is hardly human readable, especially for larger molecules. Still, its
the Chemical uniqueness makes it number one for reporting identified metabo-
Translation Service lites. SMILES and InChIs are quite long; however, the InChI keys
Bio- and Chemoinformatics Approaches for Metabolomics Data Analysis 55
}
#construct url for request
baseUrl <- "http://cts.fiehnlab.ucdavis.edu/
service/convert"
queryUrl <- paste0(baseUrl, "/", from, "/",
to, "/", queryString)
#use jsonlint fromJSON
queryResult <-
fromJSON(URLencode(queryUrl))
print(queryResult$result)
}
3.6 Manual Both two big pathway servers MetaCyc and KEGG offer web APIs
and Automated to interact with them. The KEGG database utilizes a simple REST
Mapping on KEGG web service for interaction. Different functions allow the retrieval
Pathways of information on pathways, enzymes, and their linked compounds.
This allows querying the DB within a workflow. The functionalities
include a mapper, which highlights compounds of interest with
default or user-defined colors.
1. This is a simple example how to retrieve a .png file manually
using the KEGG Search & Color function (http://www.
genome.jp/kegg/tool/map_pathway2.html). In this example,
all metabolites of the upper glycolysis pathway are colored in
red and all of the lower glycolysis pathway are in blue.
C00267 red C00074 blue
C00103 red C00036 blue
C00668 red C00022 blue
C05345 red C00186 blue
C05378 red C00024 blue
C00118 red C00033 blue
C00110 red
C00236 red
C00197 red
C00631 red
2. These values can be copied and pasted into the field “Enter
objects one per line followed by bgcolor, fgcolor:” (compound
input in Fig. 3). If pathway maps of a specific organism should
be colored, this organism has to be selected under “Search
Against” (organism input in Fig. 3). After pressing the “Exec”
button, a list of links to the colored pathway maps is available in
the browser. The number in brackets behind each pathway
name indicates the number of compounds that have been
mapped to this specific pathway.
3. Since manual mapping of several hundreds of pathways
can be tedious, an automated version would be useful. This can
be achieved by a small R script. The first example just performs
simple highlighting of compounds on a single pathway map.
58 Michael Witting
Fig. 3 Manual mapping of compounds is performed with the KEGG Mapper (http://www.genome.jp/kegg/tool/
map_pathway2.html). KEGG IDs of compounds of interest are pasted into the text field marked with compound
input; optional fore and background color can be defined. An organism can be selected by entering the respec-
tive three letter code in the field marked as organism input. After pressing the Exec button, a list with all
pathways containing minimum one of the compounds is shown. By clicking on the respective pathway, the
map is shown
4 Notes
References
1. Benton HP, Wong DM, Trauger SA et al (2008) ment, matching, and identification. Anal Chem
XCMS2: processing tandem mass spectrometry 78:779–787
data for metabolite identification and structural 3. Müller C, Dietz I, Tziotis D et al (2013)
characterization. Anal Chem 80:6382–6389 Molecular cartography in acute chlamydia
2. Smith CA, Want EJ, O'Maille G et al (2006) pneumoniae infections—a non-targeted
XCMS: processing mass spectrometry data for metabolomics approach. Anal Bioanal Chem
metabolite profiling using nonlinear peak align- 405:5119–5131
Bio- and Chemoinformatics Approaches for Metabolomics Data Analysis 61
4. Stanstrup J, Gerlich M, Dragsted LO et al phy mass spectrometry and its use for peak
(2013) Metabolite profiling and beyond: annotation in metabolomics.
approaches for the rapid processing and anno- Metabolomics:1–11
tation of human blood serum mass spectrome- 17. Peironcely JE et al (2012) OMG: Open
try data. Anal Bioanal Chem Molecule Generator. J Cheminformatics
405(15):5037–5048 4:1–13
5. Kind T, Fiehn O (2007) Seven golden rules for 18. Wolf S et al (2010) In silico fragmentation for
heuristic filtering of molecular formulas computer assisted identification of metabolite
obtained by accurate mass spectrometry. BMC mass spectra. BMC Bioinformatics 11:148
Bioinformatics 8:105 19. Gerlich M, Neumann S (2013) MetFusion:
6. Tziotis D, Hertkorn N, Schmitt-Kopplin P integration of compound identification strate-
(2011) Kendrick-analogous network visualisa- gies. J Mass Spectrom 48:291–298
tion of ion cyclotron resonance Fourier trans- 20. Kanehisa M, Goto S (2000) KEGG: Kyoto
form mass spectra: improved options for the encyclopedia of genes and genomes. Nucleic
assignment of elemental compositions and the Acids Res 28:27–30
classification of organic molecular complexity.
Eur J Mass Spectrom 17:415–421 21. Kanehisa M et al (2006) From genomics to
chemical genomics: new developments in
7. Witting M, Lucio M, Tziotis D et al (2015) KEGG. Nucleic
DI-ICR-FT-MS-based high-throughput deep Acids Res 34(suppl 1):D354–D357
metabotyping: a case study of the Caenorhabditis
Elegans–Pseudomonas Aeruginosa infection 22. Wishart DS et al (2012) HMDB 3.0—the
model. Anal Bioanal Chem 407:1059–1073 human metabolome database in 2013. Nucleic
Acids Res 41((Database issue)):D801–D807.
8. Treutler H, Neumann S (2016) Prediction, https://doi.org/10.1093/nar/gks1065
detection, and validation of isotope clusters in
mass spectrometry data. Meta 6:E37 23. Wishart DS et al (2009) HMDB: a knowledge-
base for the human metabolome. Nucleic Acids
9. Kerber A et al (1998) MOLGEN 40 Match- Res 37(Database):D603–D610
communications in mathematical and in com-
puter. Chemistry 37:205–208 24. Sud M et al (2007) LMSD: LIPID MAPS
structure database. Nucleic Acids Res 35(suppl
10. Peironcely JE et al (2012) OMG: Open 1):D527–D532
Molecule Generator. J Cheminformatics 4:21
25. Caspi R et al (2008) The MetaCyc database of
11. Jaghoori MM et al (2013) PMG: multi-core metabolic pathways and enzymes and the
Metabolite Identification. Electronic Notes in BioCyc collection of pathway/genome data-
Theoretical Computer Science 299:53–60 bases. Nucleic Acids Res 36(suppl
12. Kind T, Scholz M, Fiehn O (2009) How large 1):D623–D631
is the metabolome? A critical analysis of data 26. David S. Wishart, Yannick Djoumbou
exchange practices in chemistry. PLoS One Feunang, Ana Marcu, An Chi Guo, Kevin
4:e5440 Liang, Rosa Vázquez-Fresno, Tanvir Sajed,
13. Wohlgemuth G et al (2010) The chemical Daniel Johnson, Carin Li, Naama Karu, Zinat
translation service—a web-based tool to Sayeeda, Elvis Lo, Nazanin Assempour, Mark
improve standardization of metabolomic Berjanskii, Sandeep Singhal, David Arndt,
reports. Bioinformatics 26:2647–2648 Yonjie Liang, Hasan Badran, Jason Grant,
14. Steinbeck C et al (2003) The chemistry devel- Arnau Serra- Cayuela, Yifeng Liu, Rupa
opment kit (CDK): an open-source java library Mandal, Vanessa Neveu, Allison Pon, Craig
for chemo- and bioinformatics. J Chem Inf Knox, Michael Wilson, Claudine Manach,
Comput Sci 43:493–500 Augustin Scalbert; HMDB 4.0: the human
15. Willighagen EL, Mayfield JW, Alvarsson J, Berg metabolome database for 2018, Nucleic Acids
A, Carlsson L, Jeliazkova N, Kuhn S, Pluskal T, Research, gkx1089, https://doi.
Rojas-Chertó M, Spjuth O, Torrance G, Evelo org/10.1093/nar/gkx1089
CT, Guha R, Steinbeck C (2017) The Chemistry 27. Juty N, Le Novère N, Laibe C (2012)
Development Kit (CDK) v2.0: atom typing, Identifiers.org and MIRIAM registry: commu-
depiction, molecular formulas, and substructure nity resources to provide persistent identifica-
searching. J Cheminform 9:33. https://doi. tion. Nucleic Acids Res 40:D580–D586
org/10.1186/s13321-017-0220-4 28. Beisken S et al (2013) KNIME-CDK: work-
16. Cao M et al (2014) Predicting retention time flow-driven cheminformatics. BMC
in hydrophilic interaction liquid chromatogra- Bioinformatics 14:257
Part II
Methods
Chapter 5
Abstract
Metabolomics aims at the identification and quantification of key-end point metabolites, basically polar, in
order to study changes in biochemical activities in response to pathophysiological stimuli or genetic modi-
fications. Targeted profiling assays have enjoyed a growing popularity during the last years with LC-MS/
MS as a powerful tool for development of such (semi-) quantitative methods for a large number of metab-
olites. Here we describe a method for absolute quantification of ca. 100 metabolites belonging to key
metabolite classes such as sugars, amino acids, nucleotides, organic acids, and amines with a hydrophilic
interaction liquid chromatography (HILIC) system comprised of ultra (high) performance liquid chroma-
tography (UHPLC) with detection on a triple-quadrupole mass spectrometer operating in both positive
and negative electrospray ionization modes.
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_5, © Springer Science+Business Media, LLC, part of Springer Nature 2018
65
66 Christina Virgiliou et al.
2 Materials
3 Methods
3.1 UPLC Method With regard to UPLC conditions, the parameters for binary sol-
vent manager are as follows: flow rate is kept constant throughout
the whole analysis at 0.5 ml/min and the following gradient is
programmed: 4 min isocratic step at 100% A, then rising to 40% B
linearly over the next 21 min and finally reaching 85% B over
5 min. The column is equilibrated for 10 min in the initial condi-
tions. Regarding the sample manager, the flow through needle sys-
tem is applied in the present protocol. Injection volume is set at
5 μl and sample temperature at 6 °C. Injection system is subjected
to two washing cycles with a strong solvent and a weak solvent
prior to injection and one cycle of 6 s with the strong solvent for
post-wash. Column temperature is set to 40 °C.
3.2 MS Method In order to edit the MS method, find the optimum parameters for
the MRM transition of each metabolite. For this protocol, manual
optimization is performed in order to find precursor and product
ions and the optimum cone voltage and collision energy. With
70 Christina Virgiliou et al.
3.3 Sample Extraction of samples may vary between different matrices. So far
Preparation the described method or its variants is tested with serum, urine,
amniotic fluid, intra-/extracellular content, feces, and various types
of animal tissue but also in foods such as honey, muscle tissue, and
flour. The sample preparation procedure for blood serum samples
is presented below.
1. Allow samples to thaw at room temperature.
2. Mix 50 μl of sample with 130 μl MeCN, 10 μl H2O, and 10 μl
MeOH in an eppendorf vial of 1 ml by the use of variable vol-
ume pipette 20–200 μl (see Note 7).
Table 2
Analytes that can be detected and monitored with the HILIC-MS/MS method
Cone
Monoisotopic Precursor Product voltage Collision Rt Molecular Dwell
A/A Metabolites Formula mass ion ion (V) energy (V) Polarity (min) weight time
1 2-Hydroxyisobutyric acid C4H8O3 104.04 103 57 30 10 − 8.0 104.10 0.005
2 2-Hydroxyisovaleric acid C5H10O3 118.06 117 71 30 12 − 6.0 118.13 0.005
3 2-MethylHippuric acid C10H11NO3 193.20 192 148 35 12 − 8.2 193.20 0.005
4 3-Methylhistidine C7H11N3Ο2 169.09 170 109 30 10 + 19.0 169.18 0.003
5 4-Hydroxyphenyllactate C9H10O4 182.05 181 63 33 12 − 10.0 182.17 0.02
6 a-Ketoglutaric acid C5H6O5 146.01 145 101 20 9 − 16.0 146.11 0.02
7 Acetylcarnitine C9H17NO4 203.12 204 85 30 10 + 14.4 203.23 0.003
8 Adenine C5H5N5 135.05 136 119 40 20 + 3.6 135.13 0.003
9 Adenosine C10H13N5O4 267.10 268 136 20 15 + 4.4 267.24 0.003
10 Adipic acid C6H10O4 146.06 145 101 25 12 − 16.0 146.14 0.005
12 Alanine C3H7NO2 89.05 90 44 20 10 + 16.0 89.09 0.005
13 Arabitol C5H12O5 152.07 151 89 25 10 − 9.9 152.14 0.02
14 Arginine C6H14N4O2 174.11 175 70 30 19 + 21.9 174.20 0.005
15 Ascorbic acid C6H8O6 176.03 176 70 20 15 + 3.6 176.12 0.02
16 Asparagine C4H8N2O3 132.05 133 74 20 14 + 18.2 132.11 0.02
17 Aspartic acid C4H7NO4 133.04 134 74 18 16 + 21.8 133.11 0.02
HILIC-MS/MS Multi-Targeted Method for Metabolomics Applications
(continued)
71
Table 2
72
(continued)
Cone
Monoisotopic Precursor Product voltage Collision Rt Molecular Dwell
A/A Metabolites Formula mass ion ion (V) energy (V) Polarity (min) weight time
20 Biotin C10H16N2O3S 244.09 245 227 25 14 + 8.1 244.31 0.003
21 Cadaverine C5H14N2 102.12 103 86 15 8 + 20.4 102.17 0.003
22 Caffeine C8H10N4O2 194.08 195 138 38 18 + 0.9 194.19 0.032
Christina Virgiliou et al.
(continued)
73
Table 2
74
(continued)
Cone
Monoisotopic Precursor Product voltage Collision Rt Molecular Dwell
A/A Metabolites Formula mass ion ion (V) energy (V) Polarity (min) weight time
60 Malic acid C4H6O5 134.02 133 115 22 10 − 20.2 134.08 0.02
61 Malonic acid C3H4O4 104.01 103 59 15 9 − 14.4 104.06 0.02
62 Maltose C12H22O11 342.12 341 161 25 8 − 18.1 342.30 0.02
Christina Virgiliou et al.
(continued)
75
Table 2
76
(continued)
Cone
Monoisotopic Precursor Product voltage Collision Rt Molecular Dwell
A/A Metabolites Formula mass ion ion (V) energy (V) Polarity (min) weight time
100 Uracil C4H4N2O2 112.03 113 70 40 15 + 1.9 112.08 0.02
101 Uric acid C5H4N4O3 168.03 169 141 35 15 + 16.3 168.11 0.02
102 Uridine C9H12N2O6 244.07 243 110 35 16 − 4.7 244.12 0.005
Christina Virgiliou et al.
3.4 UPLC-MS/MS The following steps describe how to set up a sample table for data
Analysis acquisition. The following sample order has been evaluated to be
optimal for both throughput and quality controls:
1. Run a gradient without injection in order to evaluate column
performance and have a measure of minimum and maximum
pressure during analysis (see Note 11).
2. Perform six replicate injections of a standard mixture in order to
pretest systems performance (retention time, signal) and to
equilibrate system (see Note 12).
3.5 Data Treatment- Quantitation can be performed in both manual and automated
Quantification ways using vendor software. In the present protocol TargetLynx
of Metabolites (Waters) is used. A quantify method must be created before inte-
gration or quantification can be performed. Related software from
other vendors includes MultiQuant (Sciex), Xcalibur (Thermo
Fischer), and MassHunter WorkStation-Quantitative Analysis
(Agilent).
TargetLynx data can be saved as .qld files for further manipula-
tion. Complete summary of the results and .qld files (area, response,
concentration, S/N, SD, measured concentration, etc.) can be
exported as .txt files and open with excel Microsoft program for
further treatment.
In case of external calibration curve approach, once you find
the optimum method parameters, calibration of standards and
quantification can be performed directly with only one process (see
Note 14).
HILIC-MS/MS Multi-Targeted Method for Metabolomics Applications 79
4 Notes
References
1. Nicholson JK, Lindon JC, Holmes E (1999) 9. Griffiths WJ, Koal T, Wang Y et al (2010)
‘Metabonomics’: understanding the meta- Targeted metabolomics for biomarker discov-
bolic responses of living systems to patho- ery. Angew Chem Int Ed Engl 49:5426–5445
physiological stimuli via multivariate statistical 10. Michopoulos F, Whalley N, Theodoridis G
analysis of biological NMR spectroscopic data. et al (2014) Targeted profiling of polar intra-
Xenobiotica 29:1181–1189 cellular metabolites using ion-pair-high per-
2. Nicholson JK, Lindon JC (2008) Systems biol- formance liquid chromatography and -ultra
ogy: Metabonomics. Nature 455:1054–1056 high performance liquid chromatography
3. Nicholson JK, Connelly J, Lindon JC et al coupled to tandem mass spectrom. : appli-
(2002) Metabonomics: a platform for studying cations to serum, urine and tissue extracts.
drug toxicity and gene function. Nat Rev Drug J Chromatogr A 1349:60–68
Discov 1:153–161 11. Roberts LD, Souza AL, Gerszten RE et al
4. Theodoridis GA, Gika HG, Wilson ID (2011) Targeted metabolomics. Curr Protoc Mol Biol
Mass spectrometry-based holistic analytical 98:302.1–302.24
approaches for metabolite profiling in sys- 12. Gika HG, Theodoridis GA, Vrhovsek U
tems biology studies. Mass Spectrom Rev et al (2012) Quantitative profiling of polar
30:884–906 primary metabolites using hydrophilic inter-
5. Zhou B, Xiao JF, Tuli L, Ressom HW (2012) action ultrahigh performance liquid chro-
LC-MS- based metabolomics. Mol BioSyst matography-tandem mass spectrometry.
8:470–481 J Chromatogr A 1259:121–127
6. Fiehn O, Robertson D, Griffin J et al (2007) 13. Schiesel S, Lämmerhofer M, Lindner W
The metabolomics standards initiative (MSI). (2010) Multitarget quantitative metabolic
Metabolomics 3:175–178 profiling of hydrophilic metabolites in fer-
7. Theodoridis GA, Gika HG, Want EJ et al mentation broths of β-lactam antibiotics
(2012) Liquid chromatography-mass spec- production by HILIC-ESI-MS/MS. Anal
trometry based global metabolite profiling: a Bioanal Chem 396:1655–1679
review. Anal Chim Acta 711:7–16 14. Coulier L, Bas R, Jespersen S et al (2006)
8. Verpoorte R, Choi YH, Kim HK (2010) Simultaneous quantitative analysis of metabo-
Metabolomics: will it stay? Phytochem Anal lites using ion-pair liquid chromatography-
21:2–3 electrospray ionization mass spectrometry.
Anal Chem 78:6573–6582
HILIC-MS/MS Multi-Targeted Method for Metabolomics Applications 81
Abstract
A protocol for the preparation of tissue extracts for the targeted analysis of ca. 150 polar metabolites,
including those involved in central carbon metabolism is described, using a reversed-phase ion pair U(H)
PLC-MS method. Data collection enabled by multiple-reaction monitoring provides highly specific, sensi-
tive acquisition of metabolic intermediates with a wide range of physicochemical properties and pathway
coverage. Technical aspects are discussed for method transfer along with the basic principles of sample
sequence setup, data analysis, and validation. General comments are given to help the assessment of data
quality and system performance.
Key words Metabonomics, Targeted analysis, Mass spectrometry, Metabolites, Ion pair
chromatography
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_6, © Springer Science+Business Media, LLC, part of Springer Nature 2018
83
84 Filippos Michopoulos
2 Materials
2.1 Preparation For all the analytical standards listed in Table 1, the appropriate
of Analytical amount is weighed in a 1.5 mL Eppendorf tube to result in a 1 mL
Standards, Test 50 mM solution in MeOH/H2O 50/50 v/v (stock solution). For
Mixture, and Infusion amino acids and nucleotides, solubility is improved by the addition
Solution of small amounts of HCl, while other classes of metabolite may
require small amounts of 1 M NaOH. Stock solutions must be
diluted 1/100 v/v with HPLC grade water to produce a solution
(dilution A) with nominal concentration of 500 μM which will be
further diluted 1/50 with either HPLC water (dilution B) or
MeOH/H2O 50/50 v/v (infusion solution) to result final stan-
dard concentration of 10 μM. Stock solutions, dilutions A and B
should be transferred to 1.8 mL cryovials and stored at −20 °C as
reference material for future use.
An aliquot of 30 μL of the appropriate stock solution standards
(see Table 1) can be mixed in a 15 mL Falcon tube and concentra-
tion adjusted to 100μΜ with HPLC water before dividing into
10 μL aliquots (test mixture 1–6) and storage at −20 °C for use for
batch validation.
2.2.2 Mobile Phase B In a 1 L solvent bottle mix 800 mL of MeOH with 200 mL of
isopropanol and manually shake for 1 min before connecting to
chromatographic system.
2.2.3 Syringe In a 0.5 L solvent bottle, mix equal volumes of isopropanol with
and Needle Wash acetonitrile and shake to ensure mixing.
2.2.4 Seal Wash Solvent In a 0.5 L solvent bottle, mix 400 mL of HPLC water with 100 mL
of isopropanol and shake to ensure mixing (see Notes 1–3).
Table 1
List of metabolites measured with the current methodology, mass spectrometer parameters, and text
mixtures composition
(continued)
Ion Pair Chromatography for Endogenous Metabolites LC-MS Analysis in Tissue… 87
Table 1 (continued)
(continued)
88 Filippos Michopoulos
(continued)
Ion Pair Chromatography for Endogenous Metabolites LC-MS Analysis in Tissue… 89
Table 1 (continued)
(continued)
90 Filippos Michopoulos
Table 1 (continued)
(continued)
Ion Pair Chromatography for Endogenous Metabolites LC-MS Analysis in Tissue… 91
Table 1 (continued)
3 Methods
30, ion source gas1 60, and ion source gas2 50. Gas values are
arbitrary units.
3.3 Tissue Extraction Tissue samples must be processed and extracted from frozen to
and Preparation reduce endogenous metabolite degradation. For the present pro-
tocol, we can use a combined extraction homogenization approach
that is performed using a Precellys 24 system with an attached tem-
perature control unit. For soft tissue samples, we advise the use of
CK14 tubes, while for harder tissue, the CK28R format is more
appropriate. Extraction and homogenization is achieved in the
appropriate tube format from a 50 mg of frozen tissue with 1 mL
ACN/MeOH/H2O 40/40/20 v/v/v. Tubes must be shaken at
5000 rpm for 20 s, and the process must be repeated three times
with an intermittent pause of 30 s between each repeat. While the
extraction and homogenization is taking place, the extraction
chamber unit must be at the lowest possible temperature and not
more than 10 °C. A clear supernatant is obtained after centrifuga-
tion at 10691 × g for 5 min at 0 °C, and this is transferred to cryo-
vial for storage at a minimum −20 °C until analysis. The
extraction-homogenization procedure is repeated with fresh sol-
vent as described above, and the resulted clear supernatant is com-
bined in the same cryovial.
3.6 Data Analysis Raw spectrometric data are processed with MultiQuant software to
obtain peak areas for each of the detected metabolites across the
sample set. The first step prior to peak integration is the visual con-
firmation of the retention time for each metabolite peak by
comparing the trace obtained for the test mixture, spiked QC, and
the individual samples. Given the inability to obtain biological
matrix free from the endogenous metabolites being determined,
the solvent blank injection data can be used to define the back-
ground threshold values for each metabolite to ensure signals are
associated with actual metabolite presence and not the background.
Smoothing factor and peak splitting are two additional parameters
that must be customized using the sample injections in order to
obtain accurate and reproducible peak integration. The peak inte-
gration report can be exported as a text file from the MultiQuant
software and further processed in customer optimized visualization
and statistical software packages. For example, in Excel univariate
statistical analysis (f-test, t-test, ANOVA) can be performed to vali-
dated significant differences across meaningful group comparison
on metabolite level very simply. It is also essential to extend the
univariate analysis and combine with analytical reproducibility data
(coefficient of variation values, CV) to reinforce statistical signifi-
cance (see Note 8). Data normalization is also essential to reduce
bias and trends due to analysis order or loss of signal intensity since
no internal standards are used in this analysis protocol. Median
value normalization is highly regarded as an adequate approach for
this type of data set. Typical validation criteria for detecting
Ion Pair Chromatography for Endogenous Metabolites LC-MS Analysis in Tissue… 95
Fig. 1 Typical ion chromatogram of a text mixture injection obtained following the proposed chromatographic
separation
4 Notes
References
1. Bales JR et al (1998) Metabolic profiling of 8. Gika HG et al (2010) Does the mass spectrom-
body fluids by proton NMR: self-poisoning eter define the marker? A comparison of global
episodes with paracetamol (acetaminophen). metabolite profiling data generated simultane-
Magn Reson Med 6(3):300–306 ously via UPLC-MS on two different mass
2. Nicholson JK, Wilson ID (1989) High resolu- spectrometers. Anal Chem 82(19):8226–8234
tion proton magnetic resonance spectroscopy 9. Dunn WB, Ellis DI (2005) Metabolomics: cur-
of biological fluids. Prog Nucl Magn Reson rent analytical platforms and methodologies.
Spectrosc 21(4–5):449–501 TrAC Trends Anal Chem 24(4):285–294
3. Barzilai A et al (1991) Phosphate metabolites 10. Lenz EM, Wilson ID (2007) Analytical strate-
and steroid hormone receptors of benign and gies in Metabonomics. J Proteome Res
malignant breast tumors. A nuclear magnetic 6(2):443–458
resonance study. Cancer 67(11):2919–2925 11. Lindon JC, Nicholson JK (2008) Analytical
4. Gavaghan CL et al (2000) An NMR-based technologies for metabonomics and metabolo-
metabonomic approach to investigate the bio- mics, and multi-omic information recovery.
chemical consequences of genetic strain differ- TrAC Trends Anal Chem 27(3):194–204
ences: application to the C57BL10J and 12. Theodoridis G, Gika HG, Wilson ID (2008)
Alpk:ApfCD mouse. FEBS Lett LC-MS-based methodology for global metab-
484(3):169–174 olite profiling in metabonomics/metabolo-
5. Kurhanewicz J et al (1995) Citrate as an in vivo mics. TrAC Trends Anal Chem
marker to discriminate prostate cancer from 27(3):251–260
benign prostatic hyperplasia and normal pros- 13. Shulaev V (2006) Metabolomics technology
tate peripheral zone: detection via localized and bioinformatics. Brief Bioinform
proton spectroscopy. Urology 45(3):459–466 7(2):128–139
6. Lynch MJ, Nicholson JK (1997) Proton MRS 14. Smith CA et al (2006) XCMS: processing mass
of human prostatic fluid: correlations between spectrometry data for metabolite profiling
citrate, spermine, and myo-inositol levels and using nonlinear peak alignment, matching, and
changes with disease. The Prostat identification. Anal Chem 78(3):779–787
30(4):248–255 15. Xia J et al (2012) MetaboAnalyst 2.0—a com-
7. Marx A et al (1996) Determination of the prehensive server for metabolomic data analy-
fluxes in the central metabolism of sis. Nucleic Acids Res 40(W1):W127–W133
Corynebacterium glutamicum by nuclear mag- 16. Lu W, Bennett BD, Rabinowitz JD (2008)
netic resonance spectroscopy combined with Analytical strategies for LC–MS-based targeted
metabolite balancing. Biotechnol Bioeng metabolomics. J Chromatogr B
49(2):111–129 871(2):236–242
Ion Pair Chromatography for Endogenous Metabolites LC-MS Analysis in Tissue… 97
Abstract
LC-MS untargeted analysis is a valuable tool in the field of metabolic profiling (metabonomics/metabolomics),
and the applications of this technology have grown rapidly over the past decade. LC-MS offers advantages over
other analytical platforms such as speed, sensitivity, relative ease of sample preparation, and large dynamic range.
As with any analytical approach, there are still drawbacks and challenges to overcome, but advances are con-
stantly being made regarding both column chemistries and instrumentation. There are numerous untargeted
LC-MS approaches which can be used in this ever-growing research field; these can be optimized depending on
sample type and the nature of the study or biological question. Some of the main LC-MS approaches for the
untargeted analysis of biological samples will be described in detail in the following protocol.
Key words LC-MS, Untargeted, Mass spectrometry, Liquid chromatography, Metabolic profiling
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_7, © Springer Science+Business Media, LLC, part of Springer Nature 2018
99
100 Elizabeth J. Want
2 Materials
3 Methods
Fig. 1 LC-MS untargeted workflow showing the main steps of the process. These steps are described in this
protocol
to change over the analytical run, and so this mix can also be run
at the end of the sample analysis to assess these changes. For a
more detailed description, the reader is directed to Chapter 2.
Quality Control Samples: Quality control (QC) samples are the
key to successful, robust untargeted LC-MS analyses. These sam-
ples are representative of the study sample set and are usually made
by mixing small aliquots (e.g., 10–50 μL) from all study samples.
QC samples are used for (a) conditioning of the LC column and
(b) assessment of data quality. Subheading 3.3 step 9 describes the
setup for QC samples in the run. For a more detailed description,
the reader is directed to Chapter 2.
Sample Randomization: Study samples should be randomized
within a batch to avoid bias which may arise from changes in the
system over the course of the run, e.g., decrease in detector sensi-
tivity and drifts in chromatographic retention times or mass accu-
racy. Samples can be randomized using in-house scripts, online
software (https://www.randomizer.org/; https://www.random.
org/lists/), or randomized block design in the case of larger stud-
ies. For a more detailed description, the reader is directed to
Chapter 2.
3.2 Preparation Prepare a fresh stock solution of sodium formate; 0.1 mg/mL in
of LC-MS Calibration water. Add 1 mL of stock solution to 9 mL of isopropanol (IPA) to
Solution obtain a working solution of 0.01 mg/mL. This solution can be
106 Elizabeth J. Want
3.3.1 LC Gradients 1. Examples of UPLC gradients for urine, serum, and tissue sam-
ples by both RP and HILIC analysis are shown in the following
tables: Tables 1a, 1b, 1c, 1d, 1e, 1f, and 1g. The column dimen-
sions and details are also provided. Note that these gradients
will require optimization for different column chemistries,
lengths, and particle sizes.
3.4 MS Settings Example MS settings, including cone and capillary voltage, are
shown in Table 2 below. Note that these settings are for Waters
Xevo Q-ToF mass spectrometers and will require optimization for
each specific mass spectrometer. However, they can be used as
guidelines, based on an ultra-performance liquid chromatography
(UPLC) setup with a flow rate of 400–500 μL/min and a Q-ToF
mass spectrometer.
108 Elizabeth J. Want
Fig. 2 Selected portion of a BPI chromatogram showing serum QC samples overlaid. Inset is an example of
monitoring ion intensity of a specific metabolite over the QC samples
Table 1a
Example gradient for the RP analysis of urine samples
Column = UPLC Acquity HSS T3. Flow rate = 0.5 mL/min. Mobile phases; A = 0.1%
formic acid in water, B = 0.1% formic acid in acetonitrile. Taken from Ref. 21
Table 1b
Example gradient for the HILIC analysis of urine samples
Column = UPLC Acquity HILIC. Flow rate = 0.4 mL/min. Mobile phases; A = 95%
acetonitrile, 5% water + ammonium acetate (10 mM final concentration), B = 50% ace-
tonitrile, 50% water + ammonium acetate (10 mM final concentration). Taken from
reference 21
Table 1c
Example gradient for the RP analysis of plasma/serum samples
Column = UPLC Acquity HSS T3. Flow rate = 0.4 mL/min. Mobile phases; A = 0.1%
formic acid in water, B = 0.1% formic acid in methanol. Taken from reference 22
Table 1d
Example gradient for the HILIC analysis of plasma/serum samples and
aqueous tissue extracts
Flow rate = 0.4 mL/min unless otherwise stated. Column = UPLC Acquity
HILIC. Mobile phases; A = acetonitrile/water (95:5), B = acetonitrile/water (50:50).
Taken from reference 25
Table 1e
Example gradient for the RP analysis of tissue samples—aqueous extract
Column = UPLC Acquity HSS T3. Flow rate = 0.4 mL/min. Mobile phases; A = 0.1%
formic acid in water, B = 0.1% formic acid in methanol. Taken from reference 23
LC-MS Untargeted Analysis 111
Table 1f
Example gradient for the RP analysis of tissue samples—organic extract
Column = UPLC Acquity CSH. Flow rate = 0.4 mL/min. Mobile phases; A = acetoni-
trile (ACN)/water (60:40); B = isopropanol/ACN (90:10). In both mobile phases
ammonium formate was diluted to 10 mM and formic acid to 0.1%. Taken from refer-
ence 25
Table 1g
Example gradient for the HILIC analysis of tissue samples—aqueous
extracts
Column = Acquity HILIC.Temperature = 35 °C. Flow rate = 0.4 mL/min unless oth-
erwise stated. Mobile phases; A = acetonitrile (ACN)/water (95:5); B = ACN/water
(50:5). In both A and B, the concentration of ammonium acetate is 10 mM and formic
acid is present at 1%. Taken from reference 25
112 Elizabeth J. Want
Table 2
Example MS settings for positive and negative mode ESI analysis using a
Q-ToF mass spectrometer. The main parameters are shown. These will
need to be optimized for each instrument and ESI mode and are to some
extent dependent on solvent system and mobile phase flow rate
Parameter Setting
Capillary voltage 1–3 kV electrospray
(ESI)+,1–2.5kVESImode-
Cone voltage 30 V
Source temperature e.g., 120 °C
Desolvation temperature e.g., 350 °C
Cone gas flow 25 L/h
Desolvation gas flow 900 L/h
3.6 Data It is beyond the scope of this protocol to provide a detailed descrip-
Preprocessing tion of data analysis. However, the key data analysis steps pertain-
ing to LC-MS untargeted analysis for metabolic profiling studies
are described briefly in steps 1–7. There are many different com-
mercial and freeware available to perform some or all of these steps.
These are listed in the Materials section of this protocol.
1. The first step in data preprocessing of untargeted LC-MS data
is peak picking. Define chromatographic processing regions—it
may be that it is not desirable to include the solvent front, e.g.,
0–1 min, and the re-equilibration portion of the data. Some
software may allow the user to omit these regions from further
analysis.
2. This is followed by alignment of the peaks to correct for any
retention shifts that have occurred between samples during the
analysis.
3. Peaks are then integrated using peak area through a selected
algorithm depending on the software used.
4. Data is often normalized during this process. A common
approach is to use median fold change [26] to account for dif-
ferences in sample dilutions. These are particularly common in
biological samples such as urine samples.
5. QC filtering is key in untargeted LC-MS metabolic profiling
studies. This is explained in Subheading 3.7.
6. Output of metabolite feature table for multivariate analysis.
LC-MS Untargeted Analysis 113
3.7 Data Analysis Once the metabolite table has been produced, this can be exported
into appropriate software, e.g., excel for further analysis, e.g., QC
CV filtering. This will be covered in more detail in Chapter 2.
4 Notes
References
Abstract
Metabolomic studies offer a wealth of information on cells, tissues, and biofluids. The phenotype repre-
sentation through the metabolic profiling is a valuable tool for direct diagnosis, therapeutic strategies, and
system’s biology studies. Nuclear magnetic resonance (NMR) spectroscopy provides a nondestructive and
extremely reproducible method allowing simultaneous detection of a large number of known and unknown
chemical substances.
Sample collection and preparation and experimental conditions are critical for the reliability of the
subsequent analysis. The pre-analytical phase is decisive as it could generate biased spectral data misleading
the following analysis. The formulation of standard operating procedures is thus of crucial importance in
order to access meaningful samples and results. In this protocol, we provide standardized operations and
routine procedures from sample preparation to determine the measurement details for the acquisition of
NMR spectra highlighting major methodological issues.
Key words NMR spectroscopy, Metabolic profiling, Biofluids, Cell cultures, Tissues
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_8, © Springer Science+Business Media, LLC, part of Springer Nature 2018
117
118 Dimitra Benaki and Emmanuel Mikros
2 Materials
2.4 Instrumentation: 1. 600 MHz NMR spectrometer is advisable for biological sam-
NMR Equipment ples as a compromise in terms of resolution and sensitivity vs
cost.
2. Probe of 5 mm diameter with Z gradient (e.g., BBI, TXI for
Bruker Biospin).
3. Temperature control unit; N2 supply provides higher stability
compared to dry air.
4. Automatic sample changer (B-ACS60, Bruker Biospin, or
similar).
5. Software to control sample loading, temperature equilibration
and stability, matching and tuning, shimming, pulse calibration,
acquisition, and processing (ICONNMR, Bruker Biospin, or
similar).
2.5 Buffers Buffer A (50 mL): weigh 10.2 g KH2PO4, 50 mg TSP, and 6.5 mg
Preparation NaN3, add 40 mL D2O, and dilute in ultrasonic bath. Adjust the
pH to 7.4 with KOH pellets (for a volume of 50 mL around 33–35
pellets are required). Fill up with D2O to 50 mL volume and mix
very well. Aliquot and store at 4 °C.
Buffer B (500 mL): weigh 10.05 g Na2HPO4 × 7H2O (or
5.32 g Na2HPO4), 0.4 g TSP, and 0.2 g NaN3, add 380 mL ultra-
pure water, and dilute in ultrasonic bath. Adjust the pH at 7.4 with
1 M HCl. Fill volume up to 400 mL with ultrapure water. Add
100 mL D2O and mix well. Aliquot and store at 4 °C.
Check always the pH before use.
3 Methods
3.1 Sample The initial treatment step on the biological system has to be acute
Collection and effective (to stop all the enzymatic activity) in order to freeze
3.5.1 Cells
the existing state as well as to ensure system integrity and to gener-
ate reproducible results.
It is generally accepted that approximately 107 cells produce
NMR samples of the necessary quality for metabolomic studies.
Sample collection in monolayer cultures
1. Aspirate the cell medium or collect and store at −80 °C for fur-
ther analysis of the excreted metabolites (see Note 1).
2. Place the culture dishes on ice and wash twice with ice-cold PBS
to remove residual traces of culture medium.
3. Add ice-cold methanol up to the necessary volume to cover the
culture surface (2 mL for the 10-cm-diameter culture dish,
4 mL for 75 cm2 flask or adjust the volume according to the
NMR-Based Metabolic Profiling Procedures for Biofluids and Cell and Tissue Extracts 121
3.1.2 Biofluids Biofluid collection is easier than other biological samples; however,
processing and time intervals are crucial for system integrity and
consequent sample variability.
Blood collection is usually performed in prefilled vials with
anticoagulant supplements as EDTA, citrate, or heparin. EDTA
shows strong and broad resonance lines that obscure extended and
important areas of the 1H NMR spectra of the samples [6]. The
use of non-deuterated citrate can change the endogenous metabo-
lite concentration and/or saturate NMR receiver.
Plasma collection
1. Collect whole blood into heparin tubes in the morning pre-
prandially, after overnight fasting.
2. Centrifuge at 1500 × g for 10 min at 4 °C to separate blood
cells from plasma within 30 min of collection.
3. Collect the supernatant, aliquot, and store at −80 °C.
Serum collection
1. Allow blood to clot without anticoagulant on ice for 30 min.
2. Centrifuge at 1500 × g for 15 min at 4 °C.
NMR-Based Metabolic Profiling Procedures for Biofluids and Cell and Tissue Extracts 123
9. Collect the two phases and pool with the first extraction
fractions.
10. Dry the extracts overnight in a vacuum centrifuge or under a
nitrogen stream and store at −80 °C.
3.2.2 Tissue Extraction In the case of cell pellets stored at −80 °C, the samples are left
to thaw on ice, add 2 mL ice-cold methanol, vortex to disrupt the
pellet, and follow the extraction procedure described above.
3.3 NMR Sample The use of a buffer solution is necessary to ensure the stable and
Preparation constant pH of the samples. Proton resonances can be strongly
dependet on the pH and small fluctuations result in signal shifts,
hampering further analysis. The buffers used in NMR studies are
the sodium or potassium phosphates, as their capacity covers the
physiological pH range. It is advisable to generate quality control
(QC) samples by pooling 5 μL of each sample.
NMR-Based Metabolic Profiling Procedures for Biofluids and Cell and Tissue Extracts 125
3.3.1 Cell and Tissue The dried cell and tissue extracts are reconstituted in 10% buffer A
Extracts: Polar Metabolites and 90% D2O (see Note 7).
1. Prepare the necessary volume of 10% buffer A in D2O 99.9%.
2. Leave the samples to thaw at room temperature.
3. Add 650 μL of the solution (step 1), and mix properly (use
vortex and/or ultrasonic bath) to ensure the resuspension of all
the contents.
4. Centrifuge at 18,900 × g for 10 min at 4 °C.
5. Transfer 550 μL of the supernatant to a clean 5-mm-diameter
NMR tube.
3.3.2 Cell and Tissue 1. Prepare the necessary volume of CDCl3 0.03% v/v TMS solu-
Extracts: Nonpolar tion. During sample reconstitution, place the bottle on ice, and
Metabolites close the cup immediately after use to prevent, as much as pos-
sible, TMS evaporation.
2. Leave the samples to thaw at room temperature.
3. Add 650 μL of the solution (step 1), and mix properly (use
vortex and/or ultrasonic bath) to ensure the resuspension of all
the content.
4. Centrifuge at 18,900 × g for 10 min at 4 °C.
5. Transfer 550 μL of the supernatant in a clean 5-mm-diameter
NMR tube.
3.4.1 Specific Pulse 1. The proton 1D experiment, holding the major metabolomic
Sequence Parameters information (with the exception of samples with high content
of macromolecules), is acquired using the NOESY-presaturation
pulse sequence with gradients (noesygppr1d, Bruker library)
offering the optimum water suppression (see Note 12). A spec-
tral width of 20 ppm is required with a sampling of 64k points
resulting in an acquisition time of 2.7 s. The mixing time at
10 ms for the NOESY sequence is optimum in combination
with the presaturation and provides the best compromise for
water suppression and relaxation effect suppression thus affect-
ing peak quantification to a lesser extent. Plasma and serum
spectral width is adjusted to 30 ppm, and sampling is increased
up to 96k points resulting thus in an acquisition time of 2.7 s.
The 90 deg. pulse width should be optimized for each sample
separately (part of the acquisition automation routine) and kept
constant for all the spectra of the same sample. The processing
includes zero filling and exponential multiplication, phase cor-
rection, and axis calibration.
2. J-resolved pdeudo 2D experiments are very fast (5–10 min,
depending on the analysis and number of scans) and of utmost
importance offering the possibility to resolve overlapped sig-
nals. A spectral width of 16 ppm in the 1H axis, while 70–80 Hz
for the J coupling, is enough, with 12k points, 40 increments,
and 4–8 scans (4×n). Processing includes zero filling in both
dimensions (to 16 k and 256 for F2 and F1, respectively), line
broadening multiplication with a factor of 0.3 Hz, baseline cor-
rection, an additional tilt by 45 deg. step, and symmetrization
about the J 0 Hz line.
Additional spectra are required in the case of plasma and
serum samples (and generally for biofluids with proteins), either
to reduce the disturbance created by the macromolecular enti-
ties contribution (CPMG) or specific to derive the information
carried by large biomolecules (diffusion edited).
3. The T2 filter (relaxation edited) using the Carr-Purcell-
Meiboom-Gill sequence is applied to suppress the high MW
contribution (cpmgpr1d pulse sequence, Bruker library, with
128 Dimitra Benaki and Emmanuel Mikros
4 Notes
Acknowledgments
References
1. Chen R, Mias GI, Li-Pook-Than J et al (2012) 2. Dietmair S, Timmins NE, Gray PP et al (2010)
Personal omics profiling reveals dynamic Towards quantitative metabolomics of mam-
molecular and medical phenotypes. Cell malian cells: development of a metabolite
148(6):1293–1307. https://doi. extraction protocol. Anal Biochem 404:155–
org/10.1016/j.cell.2012.02.009. Cohen 164. https://doi.org/10.1016/j.
J (March, 2012). Examining his own body, ab.2010.04.031
stanford geneticist stops diabetes in its tracks. 3. Collinet H, Renault D (2012) Metabolic
News.sciencemag.org. Retrieved from http:// effects of CO2 anaesthesia in Drosophila
www.sciencemag.org/news/2012/03/ Melanogaster. Biol Lett 8:1050–1054.
examining-his-own-body-stanford-geneticist- https://doi.org/10.1098/rsbl.2012.0601
stops-diabetes-its-tracks
NMR-Based Metabolic Profiling Procedures for Biofluids and Cell and Tissue Extracts 131
4. Ghini V, Unger FT, Tenori L et al (2015) metabonomic procedures for NMR spectros-
Metabolomics profiling of pre-and post- copy of urine, plasma, serum and tissue extracts.
anesthesia plasma samples of colorectal patients Nat Protoc 2(11):2692–2703. https://doi.
obtained via Ficoll separation. Metabolomics org/10.1038/nprot.2007.376
11:1769–1778. https://doi.org/10.1007/ 9. Sapcariu SC, Kanashova T, Weindl D et al
s11306-015-0832-5 (2014) Simultaneous extraction of proteins
5. Overmyer KA, Thonusin C, Qi NR et al (2015) and metabolites from cells in culture. MethodsX
Impact of anesthesia and euthanasia on metab- 1:74–80. https://doi.org/10.1016/j.
olomics of mammalian tissues: studies in a mex.2014.07.002
C57BL/6J mouse model. PLoS One 10. Le Belle JE, Harris NG, Williams SR et al
10(2):e0117232. https://doi.org/10.1371/ (2002) A comparison of cell and tissue
journal.pone.0117232 extraction techniques using high-resolution
6. Nicholson JK, Buckingham MJ, Sadler PJ 1
H-
NMR spectroscopy. NMR Biomed
(1983) High resolution 1H NMR studies of 15:37–44
vertebrate blood and plasma. Biochem 11. Gowda NGA, Raftery D (2014) Quantitating
J 211(3):605–615 metabolites in protein precipitated serum using
7. Keun HC, Athersuch TJ (2011) Nuclear magnetic NMR spectroscopy. Anal Chem 86(11):5433–
resonance (NMR)-based metabolomics. In: Metz 5440. https://doi.org/10.1021/ac5005103
TO (ed) Metabolic profiling, Methods in molecu- 12. Kapoore RV, Coyle R, Staton CA et al (2015)
lar biology, vol vol 708. Springer Protocols, Cell line dependence of metabolite leakage in
Humana Press, New York, pp 321–334. https:// metabolome analyses of adherent normal and
doi.org/10.1007/978-1-61737-985-7 cancer cell lines. Metabolomics 11:1743–1755.
8. Beckonert O, Keun HC, Ebels TMD et al https://doi.org/10.1007/
(2007) Metabolic profiling, metabolomic and s11306-015-0833-4
Chapter 9
Abstract
Untargeted metabolomics refers to the high-throughput analysis of the metabolic state of a biological
system (e.g., tissue, biological fluid, cell culture) based on the concentration profile of all measurable free
low molecular weight metabolites. Gas chromatography-mass spectrometry (GC-MS), being a highly sen-
sitive and high-throughput analytical platform, has been proven a useful tool for untargeted studies of
primary metabolism in a variety of applications. As an omic analysis, GC-MS metabolomics is a multistep
procedure; thus, standardization of an untargeted GC-MS metabolomics protocol requires the integrated
optimization of pre-analytical, analytical, and computational steps. The main difference of GC-MS metab-
olomics compared to other metabolomics analytical platforms, including liquid chromatography-MS, is
the need for the derivatization of the metabolite extracts into volatile and thermally stable derivatives, the
latter being quantified in the metabolic profiles. This analytical step requires special care in the optimiza-
tion of the untargeted GC-MS metabolomics experimental protocol. Moreover, both the derivatization of
the original sample and the compound fragmentation that takes place in GC-MS impose specialized
GC-MS metabolomic data identification, quantification, normalization and filtering methods. In this
chapter, we describe the integrated protocol of untargeted GC-MS metabolomics with both the analytical
and computational steps, focusing on the GC-MS specific parts, and provide details on any sample depend-
ing differences.
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_9, © Springer Science+Business Media, LLC, part of Springer Nature 2018
133
134 Matthaios-Emmanouil P. Papadimitropoulos et al.
2 Materials
2.3 Wet Lab 1. Glass homogenizer with Teflon pestle (Thomas Scientific,
Equipment Swedesboro, NJ, USA).
2. Automatic pipettes.
3. Pipettes with glass tips (Drummond Scientific, Broomall, PA,
USA).
4. Water bath.
5. Cooling centrifuge (Thermo Fisher Scientific, Waltham, MA,
USA).
Untargeted GC-MS Metabolomics 137
2.4 GC-MS 1. 3800 series GC (Varian, Palo Alto, CA, USA—now Bruker,
Equipment Billerica, MA, USA).
and Consumables 2. Saturn 2200 series MS ion trap (Varian, Palo Alto, CA, USA—
now Agilent, Santa Clara, CA, USA)—the protocol provided
can be applied to any GC-MS equipment.
3. CP-8410 autosampler (Bruker, Billerica, MA, USA).
4. GC capillary column: Zebron, ZB-50, 30 m × 0.25 mm
ID × 0.25 μm (Phenomenex, Torrance, CA, USA).
5. Helium 99.999% carrier gas (Air Liquide Hellas, Athens,
Greece).
3 Methods
3.1 Analytical 1. (In the case of tissue or cell pellets available in large quantity, if
Protocol this step is not carried out before quenching)
Intact or Lyophilized Weigh the intact tissue [11, 12] or the selected amount of
Tissues or Cell Pellets lyophilized tissue [8–10] or cell pellet (if a large quantity is
available, see industrial cell culture application [13]) rapidly
3.1.1 Extraction of Polar before it thaws; the weight is required for the addition of the
Metabolites from appropriate amount of methanol/water and internal standards
below.
(In the case of cell pellet from a certain culture volume, e.g.,
from one petri dish culture—small quantity available)
138 Matthaios-Emmanouil P. Papadimitropoulos et al.
Blood Plasma or Other 1. Add 200 μL of blood plasma into a 2 mL Eppendorf tube con-
Liquid (e.g., Culture taining 0.6 mL of ice-cold methanol and 2 μg of adonitol/
Medium) Samples ribitol and 4 μg of U-13C-d-glucose as internal standards (see
Note 3).
Untargeted GC-MS Metabolomics 139
3.1.2 TMS Derivatization 1. Vacuum dry each stored at 4 °C vial for 30 min to remove any
remaining humidity (see Note 4).
2. Add 50 μL of 20 mg/mL solution of methoxylamine hydro-
chloride in pyridine (see Notes 5 and 6) using glass pipette tips,
and mix gently (see Notes 4 and 6); the volume of the solution
to be added may vary from 30 μL for very small amounts of dry
extracts to 150 μL for larger samples—preliminary experiments
should be carried out to determine the optimal volume for a
particular sample type balancing between the need for in excess
availability of the derivatization reagents and avoiding any large
dilution of the dry extract [2, 6].
3. Incubate in a shaker incubator at 40 °C for 90 min.
4. Add MSTFA at twice the volume of the methoxylamine hydro-
chloride solution added in step 2 using glass pipette tips and
mix gently.
5. Incubate at 40 °C for at least 6 h. This is the derivatization time
at which all metabolites in a sample are estimated to have been
fully transformed into at least one of their TMS derivatives [2,
6]. This time may vary between biological systems and could be
optimized through a preliminary experiment at which the met-
abolic profile of a certain system is measured multiple times
from 15 min to 10 h of derivatization duration [2]. However,
based on our group’s experience with various systems, 6 h of
derivatization could be considered a “universal” time for most
biological sample types for the standardization of this step of
the protocol.
6. Place the vial on the autosampler for the sample to be used for
metabolic profile acquisition.
Significant note: In an optimized GC-MS metabolic profile
acquisition protocol, taking into consideration the constraint of
step 5, four [4] samples can be quantified per day with three
repetitions per sample and three runs of the solvent (pyridine) to
clean the column between samples (see the acquisition section
below). In this optimized protocol, the incubation time of step 5
is equal to 9 h.
3.1.3 GC-MS Metabolic 1. Set the injector temperature to 230 °C and the detector transfer
Profile Acquisition line, trap, and manifold temperatures to 250 °C, 220 °C, and
70 °C, respectively. These parameters can be appropriately
adapted in a GC-quadrupole MS.
140 Matthaios-Emmanouil P. Papadimitropoulos et al.
Fig. 1 Total ion count MS-reconstructed gas chromatograms for polar metabolite extracts (left panel) and the
respective mass spectrum of the lactate 2TMS derivative (right panel) of samples from (a) industrial-scale BHK
cell culture [14], (b) C57BL/6 J male mouse cortex, and (c) tomato leaf. The straight arrow shows the peak of
the lactate 2TMS derivative in each chromatogram. The metabolites corresponding to some major peaks are
also denoted. In all three samples, the internal standard ribitol has been added in the same relative concentra-
tion with respect to the sample weight (see text)
3.2.2 Data Normalization 1. Investigate whether all profiles were acquired at the same ana-
and Filtering lytical process conditions by estimating the ratio of the two
methoxime peaks of [U-13C]—glucose used as internal stan-
dard. If available, the ratio of the two methoxime peaks of
142 Matthaios-Emmanouil P. Papadimitropoulos et al.
B
11 1
10
0.9
9
with respect to internal standard ribitol
0.8
8
Relative Peak Area (RPA)
7 0.7
6
0.6
5
4 0.5
3
0.4
2 Aspartate_3TMS
Aspartate effective 0.3
1
Aspartate_2TMS
0 0.2
9 11 13 15 17 19 21
Fig. 2 (a) The algorithm for the estimation of the weights for the derivative peak areas of an amine group
containing metabolite required for the estimation of its effective peak area in each sample (see equation at the
bottom) [2, 6]; (b) the profile of the relative peak areas (RPAs) of the two derivatives and of the estimated effec-
tive RPA of aspartate over ribitol (internal standard) in a Balbc/J male mouse cortex sample over eight [8]
different derivative durations. In (a), the algorithm is shown for a metabolite with three [3] derivatives (derivi)
and a normalization sample, the profile of which was acquired at five different derivatization durations (ti)—the
algorithm should be appropriately adjusted for different conditions. The “b” constant is defined in the same
order of magnitude either of the largest derivative or the sum of derivatives (if different) in the investigated
profiles, as explained in [2, 6]
144 Matthaios-Emmanouil P. Papadimitropoulos et al.
3.2.3 Multivariate 1. The final metabolite profile dataset after normalization and fil-
Statistical Analysis tering can be used for further analysis to extract biologically
and Metabolic Network relevant conclusions. As for other omic profiles, multivariate
Reconstruction statistical analysis methods can be applied to identify correla-
tions between metabolites and/or physiological conditions.
Specifically, supervised or unsupervised clustering algorithms
can group metabolites and/or samples of similar profiles based
on a variety of profile distance metrics (e.g., hierarchical clus-
tering analysis (HCL) [19]). Moreover, methods like principal
component analysis (PCA) [20] or partial least squares (PLS)
regression [21] can be used to lower the dimensionality of the
problem, enabling the visualization of the differences between
the profiles in a 3-D space, providing also information about
the metabolites that contribute significantly to these differ-
ences. The metabolites with significantly differential concen-
tration between sets of metabolic profiles can be identified
with the multivariate significance analysis algorithm, called
significance analysis of microarrays (SAM) [22]. SAM has been
appropriately tuned for omic data, since it does not require
that they follow a particular distribution (as the t-test or F-test)
and estimated the false discovery rate for each threshold of
significance.
There exist many software platforms, publicly or commer-
cially available, which incorporate many of these bioinformatics
and data mining algorithms, some built around an interface that
Untargeted GC-MS Metabolomics 145
4 Notes
References
1. Fiehn O (2002) Metabolomics–the link samples from mice on low- and high-fat diets.
between genotypes and phenotypes. Plant Mol J Chromatogr B 879(17–18):1467–1475.
Biol 48(1–2):155–171. https://doi. https://doi.org/10.1016/j.
org/10.1007/978-94-010-0448-0_11 jchromb.2011.01.028
2. Kanani H, Chrysanthopoulos PK, Klapa MI 6. Kanani HH, Klapa MI (2007) Data correction
(2008) Standardizing GC-MS metabolomics. strategy for metabolomics analysis using gas
J Chromatogr B 871(2):191–201. https:// chromatography-mass spectrometry. Metab
doi.org/10.1016/j.jchromb.2008.04.049 Eng 9(1):39–51. https://doi.org/10.1016/j.
3. Patti GJ, Yanes O, Siuzdak G (2012) ymben.2006.08.001
Innovation: metabolomics: the apogee of the 7. Maga-Nteve C, Klapa MI (2016) Streamlining
omics trilogy. Nat Rev Mol Cell Biol GC-MS metabolomic analysis using the
13(4):263–269. https://doi.org/10.1038/ M-IOLITE software suite. IFAC-
nrm3314 PapersOnLine 49(26):286–288. https://doi.
4. Vasilopoulou CG, Margarity M, Klapa MI org/10.1016/j.ifacol.2016.12.140
(2016) Metabolomic analysis in brain research: 8. Dutta B, Kanani H, Quackenbush J, Klapa MI
opportunities and challenges. Front Physiol (2009) Time-series integrated “omic” analyses
7:183. https://doi.org/10.3389/ to elucidate short-term stress-induced
fphys.2016.00183 responses in plant liquid cultures. Biotechnol
5. Spagou K, Theodoridis G, Wilson I et al (2011) Bioeng 102(1):264–279. https://doi.
A GC-MS metabolic profiling study of plasma org/10.1002/Bit.22036
Untargeted GC-MS Metabolomics 147
9. Kanani H, Dutta B, Klapa MI (2010) Individual early indications for predisposition to latent
vs. combinatorial effect of elevated CO2 condi- insulin resistance in children conceived by
tions and salinity stress on Arabidopsis thaliana ICSI. PLoS One 9(4):e94001. https://doi.
liquid cultures: comparing the early molecular org/10.1371/journal.pone.0094001
response using time-series transcriptomic and 16. Saeed AI, Bhagabati NK, Braisted JC et al
metabolomic analyses. BMC Syst Biol 4:177. (2006) TM4 microarray software suite.
https://doi.org/10.1186/1752-0509-4-177 Methods Enzymol 411:134–193. https://doi.
10. Tooulakou G, Giannopoulos A, Nikolopoulos org/10.1016/S0076-6879(06)11009-5
D et al (2016) “Alarm photosynthesis”: cal- 17. Saeed AI, Sharov V, White J et al (2003) TM4:
cium oxalate crystals as an internal CO2 source a free, open-source system for microarray data
in plants. Plant Physiol 171(4):2577–2585. management and analysis. BioTechniques
https://doi.org/10.1104/pp.16.00111 34(2):374–378
11. Constantinou C, Chrysanthopoulos PK, 18. Allwood JW, Erban A, de Koning S et al (2009)
Margarity M, Klapa MI (2011) GC-MS metab- Inter-laboratory reproducibility of fast gas
olomic analysis reveals significant alterations in chromatography-electron impact-time of flight
cerebellar metabolic physiology in a mouse mass spectrometry (GC-EI-TOF/MS) based
model of adult onset hypothyroidism. plant metabolomics. Metabolomics 5(4):479–
J Proteome Res 10(2):869–879. https://doi. 496. https://doi.org/10.1007/
org/10.1021/pr100699m s11306-009-0169-z
12. Maga-Nteve C, Vasilopoulou CG, 19. Eisen MB, Spellman PT, Brown PO, Botstein
Constantinou C et al (2017) Sex-comparative D (1998) Cluster analysis and display of
study of mouse cerebellum physiology under genome- wide expression patterns. Proc Natl
adult-onset hypothyroidism: the significance of Acad Sci U S A 95(25):14863–14868. https://
GC-MS metabolomic data normalization in doi.org/10.1073/pnas.95.25.14863
meta-analysis. J Chromatogr B 1041- 20. Raychaudhuri S, Stuart JM, Altman RB (2000)
1042:158–166. https://doi.org/10.1016/j. Principal components analysis to summarize
jchromb.2016.12.016 microarray experiments: application to sporula-
13. Chrysanthopoulos PK, Goudar CT, Klapa MI tion time series. Pac Symp Biocomput
(2010) Metabolomics for high- resolution 2000:455–466
monitoring of the cellular physiological state in 21. Maitra S, Yan J (2008) Principle component
cell culture engineering. Metab Eng analysis and partial least squares: two dimen-
12(3):212–222. https://doi.org/10.1016/j. sion reduction techniques for regression. In:
ymben.2009.11.001 2008 Casualty actuarial society discussion
14. Vernardis SI, Goudar CT, Klapa MI (2013) paper program–applying multivariate statistical
Metabolic profiling reveals that time related models. Casualty Actuarial Society, Quebec,
physiological changes in mammalian cell perfu- pp 79–90
sion cultures are bioreactor scale independent. 22. Tusher VG, Tibshirani R, Chu G (2001)
Metab Eng 19:1–9. https://doi. Significance analysis of microarrays applied to
org/10.1016/j.ymben.2013.04.005 the ionizing radiation response. Proc Natl Acad
15. Gkourogianni A, Kosteria I, Telonis AG et al Sci U S A 98(9):5116–5121. https://doi.
(2014) Plasma metabolomic profiling suggests org/10.1073/pnas.091062498
Chapter 10
Abstract
Fecal metabolomics-based analysis indisputably constitutes a very useful tool for elucidating the biochem-
istry of digestion and absorption of the gastrointestinal system. Fecal samples represent the most suitable,
non-invasive, specimen for the study of the symbiotic relationship between the host and the intestinal
microbiota.
It is well established that the balance of the intestinal microbiota changes in response to some stimuli,
physiological such as gender, age, diet, exercise and pathological such as gastrointestinal and hepatic dis-
ease. Fecal samples have been analyzed using the most widespread analytical techniques, namely, NMR
spectroscopy, GC-MS, and LC-MS/MS. Rat fecal sample is a frequently used and particularly useful sub-
strate for metabolomics-based studies in related fields. The complexity and diversity of the nature of fecal
samples require careful and skillful handling for the effective quantitative extraction of the metabolites
while avoiding their deterioration. Parameters such as the fecal sample weight to extraction solvent vol-
ume, the nature and the pH value of the extraction solvent, and the homogenization process are some
important factors for the optimal extraction of samples, in order to obtain high-quality metabolic finger-
prints, using either untargeted or targeted metabolomics.
Key words Metabolomics, Sample preparation, Fecal samples, Rats, NMR, GC-MS, LC-MS/MS,
Fecal extract
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_10, © Springer Science+Business Media, LLC, part of Springer Nature 2018
149
150 Olga Deda et al.
2 Materials
2.3 Instrumentation 1. 500 MHz (or higher) NMR spectrometer equipped with a
Software 5 mm triple resonance probe at 300 K (or similar)—the appro-
priate software to control acquisition of fecal sample 1H NMR
spectra (matching, tuning, shimming), to set pulse calibration
parameters, and to process.
2. Agilent 7890A GC coupled to a 5975C inert XL EI/CI MSD
with triple-axis detector MS and a CTC-CH 4222 autosampler
with an Agilent HP-5ms (29 m × 250 μm × 0.25 μm) in split/
splitless mode. MSD ChemStation (Agilent Technologies,
California, USA) to acquire and process GC-MS data.
3. ACQUITY UPLC coupled to a Xevo TQD MS system (Waters,
Massachusetts, USA) with an ACQUITY HILIC, BEH amide
column (2.1 × 150 mm, 1.7 μm). Waters MassLynx® software
to collect and process LC-MS/MS data.
Rat Fecal Metabolomics-Based Analysis 153
3 Methods
Fig. 1 Rat fecal sample preparation protocols for metabolomics-based analysis using NMR spectroscopy,
GC-MS, and LC-MS/MS
4 Notes
References
1. Guinane CM, Cotter PD (2013) Role of the gut ing of grapes: solvent extraction protocol opti-
microbiota in health and chronic gastrointestinal misation. Metabolomics 8(2):175–185
disease: understanding a hidden metabolic 5. Gika H, Theodoridis G (2011) Sample prepa-
organ. Ther Adv Gastroenterol 6(4):295–308 ration prior to the LC-MS-based metabolo-
2. Holmes E, Li JV, Athanasiou T, Ashrafian H, mics/metabonomics of blood-derived samples.
Nicholson JK (2011) Understanding the role Bioanalysis 3(14):1647–1661
of gut microbiome-host metabolic signal dis- 6. Gika HG, Wilson ID, Theodoridis GA (2014)
ruption in health and disease. Trends Microbiol LC–MS-based holistic metabolic profiling.
19(7):349–359 Problems, limitations, advantages, and future
3. Deda O, Gika HG, Wilson ID, Theodoridis perspectives. J Chromatogr B 966:1–6
GA (2015) An overview of fecal sample prepa- 7. Bollard ME, Stanley EG, Lindon JC et al
ration for global metabolic profiling. J Pharm (2005) NMR-based metabonomic approaches
Biomed Anal 113:137–150 for evaluating physiological influences on bio-
4. Theodoridis G, Gika H, Franceschi P et al fluid composition. NMR Biomed
(2012) LC-MS based global metabolite profil- 18(3):143–162
Rat Fecal Metabolomics-Based Analysis 157
8. Deda O, Chatziioannou AC, Fasoula S et al 14. Bezabeh T, Somorjai RL, Smith IC (2009)
(2017) Sample preparation optimization in ICP MR metabolomics of fecal extracts: appli-
fecal metabolic profiling. J Chromatogr B cations in the study of bowel diseases. Magn
1047:115–123 Reson Chem 47(S1):S54–S61
9. Hooper LV, Midtvedt T, Gordon JI (2002) 15. Monleon D, Garcia-Valles R, Morales JM et al
How host-microbial interactions shape the (2014) Metabolomic analysis of long-term spon-
nutrient environment of the mammalian intes- taneous exercise in mice suggests increased lipoly-
tine. Annu Rev Nutr 22:283–307 sis and altered glucose metabolism when animals
10. Lamichhane S, Yde CC, Schmedes MS et al are at rest. J Appl Physiol 117(10):1110–1119
(2015) Strategy for nuclear-magnetic-16. Gao X, Pujos-Guillot E, Martin J-F et al (2009)
resonance-based metabolomics of human feces. Metabolite analysis of human fecal water by gas
Anal Chem 87(12):5930–5937 chromatography/mass spectrometry with ethyl
11. Wu J, An Y, Yao J, Wang Y, Tang H (2010) An chloroformate derivatization. Anal Biochem
optimised sample preparation method for 393(2):163–175
NMR-based faecal metabonomic analysis. 17. Gao X, Pujos-Guillot E, Sébédio J-L (2010)
Analyst 135(5):1023–1030 Development of a quantitative metabolomic
12. Saric J, Wang Y, Li J et al (2008) Species varia- approach to study clinical human fecal water
tion in the fecal metabolome gives insight into metabolome based on trimethylsilylation
differential gastrointestinal function. derivatization and GC/MS analysis. Anal
J Proteome Res 7(1):352–360 Chem 82(15):6447–6456
13. Deda O, Gika H, Panagoulis T et al (2017) 18. Virgiliou C, Sampsonidis I, Gika HG, Raikos N,
Impact of exercise on fecal and cecal metabo- Theodoridis GA (2015) Development and vali-
lome over aging: a longitudinal study in rats. dation of a HILIC-MS/MS multitargeted
Bioanalysis 9(1):21–36 method for metabolomics applications.
Electrophoresis 36(18):2215–2225
Chapter 11
Abstract
A simple analytical workflow is described for gas chromatographic-mass spectrometry (GC-MS)-based
metabolomic profiling of protic metabolites, particularly amino-carboxylic species in biological matrices.
The sample preparation is carried out directly in aqueous samples and uses simultaneous in situ heptafluo-
robutyl chloroformate (HFBCF) derivatization and dispersive liquid-liquid microextraction (DLLME),
followed by GC-MS analysis in single-ion monitoring (SIM) mode. The protocol involves ten simple
pipetting steps and provides quantitative analysis of 132 metabolites by using two internal standards. A
comment on each analytical step and explaining notes are provided with particular attention to the GC-MS
analysis of 112 physiological metabolites in human urine.
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_11, © Springer Science+Business Media, LLC, part of Springer Nature 2018
159
160 Petr Hušek et al.
Fig. 1 Reaction scheme for the threonine protic functional groups with the HFBCF reagent. The carboxyl group
yields a HFB ester and the amino group a corresponding HFB carbamate, while the hydroxyl is transformed into
a HFB carbonate. MM = Monoisotopic mass
Fig. 2 The workflow for the GC-MS metabolomic analysis of protic metabolites in aqueous biological
matrices
Fig. 3 A view on a 6 × 50 mm culture tube containing an aqueous sample (here urine) or an aqueous sample
extract and gradually added media during the sample preparation process: (a) an aqueous sample; (b) an aris-
ing two-phase system after performing steps 2–5, before the reaction initiation; (c) a turbid upper phase after
the first addition of the catalytic medium, step 6; occasionally visible CO2 bubbles can appear; (d) the organic
upper phase is clarified after step 7, which indicates that the reaction was completed; (e) increasing the
sample extract volume and its acidification in steps 8–9 enables an easy organic upper phase into an autos-
ampler vial in step 10 and final GC-MS analysis
2 Materials
2.1 Samples Samples containing no or little protein and cell residues (urine, cell
culture media) or cell and tissue extracts.
eflon-
T capped, well-tightened 4 mL glass vial. Store in a
refrigerator, where the mixture remains stable for several
months.
9. The catalytic medium: mix 1 M NaOH with pyridine in a vol-
ume ratio of 24:1 (v/v).
10. The artificial urine solution: prepare the following chemicals
in DI water to final 10 g/L urea, 1 g/L creatinine, 7 g/L
NaCl, and 3 g/L K2SO4 [12].
11. The certified urine standard (product ORG-01) containing
diagnostic organic acids (ERNDIM Foundation (http://cms.
erndimqa.nl/). For the analyte concentrations, refer to the
website. Order the latest available batch.
Diagnostic Stock
GC ions Internal L3 solution Metabolite database coding
The profiled
No. metabolites as the RT MM m/ c Stock
No. [12]a Traditional name HFBCF derivatives (min) product zq1 m/zq2 Standardb (μM)c solution c (mM) HMDB PubChem KEGG
1 108 1-Methylhistidine 1-Methylhistidine 27.2 577.1 95 350 hF 40 0.1 M HCl 10 HMDB00001 92105 C01152
2 101 2,4-Diamino 2,4-Diamino 24.74 752.0 282 256 hF 40 DI water 10 HMDB02362 470
butyric acid butyrate
3 36 2-Aminobutyric 2-Amino 13.61 511.0 284 84 hF 20 DI water 10 HMDB00452 80283 C02356
acid butyrate
4 99 2-Amino 2-Aminopimelate 24.6 765.1 338 138 hF 40 DI water 10 HMDB34252 101122
heptanedioic-
acid
5 27 2-Aminoiso- 2-Aminoiso 12.14 525.1 284 241 hF 40 DI water 50 HMDB01906 6119 C03665
butyric acid butyrate
6 22 2-Hydroxy-3- 2-Hydroxy-3- 11.16 526.0 55 299 PB 20 DI water 10 HMDB00407 99823
methylbutyric methylbutyrate
acid (isovalerate)
7 34 2-Hydroxy-3- 2-Hydroxy-3- 13.06 540.1 284 484 PB 20 DI water 10 HMDB00317 10796774
methylpentanoic methylvalerate
acid
Diagnostic Stock
GC ions Internal L3 solution Metabolite database coding
The profiled
No. metabolites as the RT MM m/ c Stock
No. [12]a Traditional name HFBCF derivatives (min) product zq1 m/zq2 Standardb (μM)c solution c (mM) HMDB PubChem KEGG
8 19 2-Hydroxybutyric 2-Hydroxybutyrate 10.49 512.0 285 241 PB 20 DI water 10 HMDB00008 11266 C05984
acid
9 112 2-Hydroxy 2-Hydroxysebacate 27.34 808.1 95 381 PB 20 Acetonitrile 10 HMDB00424 128458
decanedioic acid
10 65 2-Hydroxyglutaric 2-Hydroxyglutarate 18.34 738.0 283 239 PB 100 DI water 100 HMDB02307 439340 C00894
acid
11 11 2-Hydroxyisobutyric 2-Hydroxyisobutyrate 8.74 512.0 241 285 PB 40 DI water 10 HMDB00729 11671
acid
12 82 2-Hydroxyphenylacetic 2-Hydroxyphenylacetate 20.52 560.0 91 333 PB 20 DI water 50 HMDB00669 11970 C05852
acid
13 26 2-Hydroxyvaleric 2-Hydroxyvalerate 12.12 526.0 55 299 PB 20 DI water 10 HMDB01863 98009
acid
14 3 2-Ketoisovaleric 2-Ketoisovalerate 5.81 298.0 71 113 PB 20 DI water 10 HMDB00019 49 C00141
acid
15 68a 2-Methylcitric 2-Methylcitrate-4 18.46 552.0 152 334 PB 40 DI water 10 HMDB00379 515
acid (lactone)
16 46 3-Aminoisobutanoic 3-Aminoisobutyrate 15.02 511.0 256 112 hF 100 0.1 M HCl 100 HMDB03911 64956 C05145
acid
17 67 3-Hydroxyadipic 3-Hydroxyadipate 18.41 752.0 85 127 PB 100 0.1 M HCl 50 HMDB00345 151913
acid
18 24a 3-Hydroxybutyric 3-Hydroxybutyrate-2 11.93 512.0 268 69 PB 100 DI water 10 HMDB00357 441 C01089
acid
19 4a 3-Hydroxyisovaleric 3-Hydroxyisovalerate- 6.46 300.1 59 85 PB 100 DI water 10 HMDB00754 69362
acid 1(OH)
(continued)
Table 1
(continued)
Diagnostic Stock
GC ions Internal L3 solution Metabolite database coding
The profiled
No. metabolites as the RT MM m/ c Stock
No. [12]a Traditional name HFBCF derivatives (min) product zq1 m/zq2 Standardb (μM)c solution c (mM) HMDB PubChem KEGG
20 41 3-Hydroxy 3-Hydroxy-3-methyl 14.37 752.0 85 285 PB 100 DI water 10 HMDB00355 1662 C03761
methylglutaric glutarate
acid
21 91 3-Hydroxy 3-Hydroxyphenyl 22.61 560.0 333 277 PB 20 0.1 M HCl 100 HMDB00440 12122 C05593
phenylacetic acid acetate
22 89a 3-Hydroxyproline 3-Hydroxyproline-2 21.9 765.0 521 538 hF 20 0.1 M HCl 10 HMDB02113 11137200 C04397
23 79a 3-Hydroxysebacic 3-Hydroxysebacate- 20.01 582.1 71 271 PB 20 DI water 10 HMDB00350 3017884
acid 1(OH)
24 9a 3-Hydroxyvaleric 3-Hydroxyvalerate- 8.21 300.1 71 271 PB 40 DI water 10 HMDB00531 107802
acid 1(OH)
25 131 3-Methoxytyramine 3-Methoxytyramine 32.36 845.1 319 376 hF 20 0.1 M HCl 10 HMDB00022 1669 C05587
26 5 3-Methyl-2- 2-Keto-3- 7.34 312.1 57 85 PB 20 DI water 10 HMDB00491 47 C03465
Oxovaleric acid methylvalerate
27 47 3-Methyladipic acid 3-Methyladipate 15.53 524.1 325 55 PB 20 DI water 50 HMDB00555 6999745
28 76 3-Methylcrotonyl Methylcrotonyl 19.54 339.1 83 82 hF 40 DI water 10 HMDB00459 169485
glycine glycine
29 116 3-Methylhistidine 3-Methylhistidine 28.38 577.1 95 150 hF 40 0.1 M HCl 10 HMDB00479 64969 C01152
30 111 4-Aminobenzoic 4-Aminobenzoate 27.29 545.0 146 345 hF 20 Ethanol 10 HMDB01392 978 C00568
acid
Diagnostic Stock
GC ions Internal L3 solution Metabolite database coding
The profiled
No. metabolites as the RT MM m/ c Stock
No. [12]a Traditional name HFBCF derivatives (min) product zq1 m/zq2 Standardb (μM)c solution c (mM) HMDB PubChem KEGG
31 87 4-Hydroxybenzoic 4-Hydroxybenzoate 21.41 546.0 303 347 PB 20 Ethanol 100 HMDB00500 135 C00156
acid
32 38 4-Hydroxybutyric 4-Hydroxybutyrate 14.05 512.0 227 269 PB 100 DI water 100 HMDB00710 10413 C00989
acid
33 115 4-Hydroxycinnamic 4-Hydroxycinnamate 27.98 572.0 329 572 PB 20 DI water 10 HMDB02035 637542 C00811
acid
34 110 4-Hydroxymandelic 4-Hydroxymandelate 27.25 802.0 575 347 PB 20 DI water 10 HMDB00822 328 C11527
acid
35 96 4-Hydroxy 4-Hydroxy 23.31 560.0 289 333 PB 40 0.1 M HCl 100 HMDB00020 127 C00642
phenylacetic acid phenylacetate
36 94a 4-Hydroxyproline 4-Hydroxyproline-2 23.12 765.0 294 521 hF 20 0.1 M HCl 50 HMDB06055 69248 C01015
37 63 4-Phenylbutyric 4-Phenylbutyrate 17.95 346.1 104 147 0.1 M 100
acid (4 PB, I.S.) NaHCO3
38 97 5-Aminolevulinic 5-Aminolevulinate 23.36 539.0 283 256 hF 40 0.1 M HCl 10 HMDB01149 137 C00430
acid
39 81 5-Aminopentanoic 5-Aminovalerate 20.41 525.1 256 269 hF 40 0.1 M HCl 10 HMDB03355 138 C00431
acid
40 53 5-Hydroxyhexanoic 5-Hydroxyhexanoate 16.67 540.1 227 113 PB 100 DI water 10 HMDB00525 170748
acid
41 144 5-Hydroxyindo 5-Hydroxyin 35.16 599.0 372 599 hF 20 DI water 10 HMDB00763 1826 C05635
leacetic acid doleacetate
42 138b 5-Hydroxylysine 5-Hydroxylysine 34.12 1022.1 269 256 hF 40 0.1 M HCl 50 HMDB00450 3032849 C16741
(isomers)
(continued)
Table 1
(continued)
Diagnostic Stock
GC ions Internal L3 solution Metabolite database coding
The profiled
No. metabolites as the RT MM m/ c Stock
No. [12]a Traditional name HFBCF derivatives (min) product zq1 m/zq2 Standardb (μM)c solution c (mM) HMDB PubChem KEGG
43 37a Acetylglycine N-Acetylglycine- 13.88 525.0 256 483 hF 200 DI water 10 HMDB00532 10972
2(NH)
44 54 Aconitic Aconitate 16.78 720.0 321 492 PB 200 DI water 100 HMDB00958 444212 C02341
acid(trans)
45 45 Adipic acid Adipate 14.95 510.1 282 311 PB 40 Ethanol 100 HMDB00448 196 C06104
46 29 Alanine Alanine 12.35 497.0 270 70 hF 40 0.1 M HCl 100 HMDB00161 5950 C00041
47 93 Aminoadipic 2-Aminoadipate 22.88 751.0 124 282 hF 40 0.1 M HCl 100 HMDB00510 469 C00956
acid
48 70 Asparagine Asparagine 19.1 522.0 295 95 hF 100 0.1 M HCl 100 HMDB00168 6267 C00152
49 64 Aspartic acid Aspartate 18.3 723.0 254 496 hF 40 0.1 M HCl 100 HMDB00191 5960 C00049
50 86 Azelaic Azelaate 21.23 552.1 353 152 PB 20 Ethanol 10 HMDB00784 2266 C08261
acid
51 20 Benzoic Benzoate 10.74 304.0 105 304 PB 40 Ethanol 100 HMDB01870 243 C00180
acid
52 44 Beta-alanine 3-alanine 14.91 497.0 270 113 hF 40 0.1 M HCl 10 HMDB00056 239 C00099
53 16 Citraconic Citraconate 10.29 494.0 295 267 PB 40 DI water 10 HMDB00634 643798 C02226
acid
54 21a Citramalic Citramalate-1 10.8 312.0 85 285 PB 100 DI water 10 HMDB00426 1081 C00815
acid (lactone)
55 60a Citric acid Citrate-2 (OH) 17.79 738.0 311 269 PB 400 DI water 100 HMDB00094 311 C00158
Diagnostic Stock
GC ions Internal L3 solution Metabolite database coding
The profiled
No. metabolites as the RT MM m/ c Stock
No. [12]a Traditional name HFBCF derivatives (min) product zq1 m/zq2 Standardb (μM)c solution c (mM) HMDB PubChem KEGG
56 132 Cystathionine Cystathionine 32.42 1038.0 328 282 hF 40 0.1 M HCl 100 HMDB00099 439258 C02291
57 90 Cysteine Cysteine (total)** 22.22 755.0 328 285 hF 100 0.1 M HCl 100 HMDB00574 5862 C00097
58 124a Diaminopimelic 2,6-Diaminopimelate 29.45 1006.1 308 536 hF 40 DI water 10 HMDB01370 439283 C00666
acid
59 135 DOPA 3,4-Dihydroxy 32.63 1057.0 149 388 hF 40 0.1 M HCl 10 HMDB00609 836 C00355
phenyl alanine
60 12 Ethylmalonic Ethylmalonate 8.78 496.0 297 468 PB 40 DI water 10 HMDB00622 11756
acid
61 130 Ferulic acid 4-Hydroxy-3- 30.64 602.0 602 375 PB 20 Ethanol 100 HMDB00954 445858 C01494
(trans) methoxycinnamate
62 15 Fumaric acid Fumarate 9.53 480.0 281 253 PB 40 DI water 10 HMDB00134 444972 C00122
63 83 Glutamic acid Glutamate 20.82 737.0 310 282 hF 40 0.1 M HCl 10 HMDB03339 23327 C00217
64 107 Glutamine Glutamine 26.64 554.1 84 282 hF 100 DI water 100 HMDB00641 5961 C00064
65 30 Glutaric acid Glutarate 12.51 496.0 227 297 PB 40 DI water 10 HMDB00661 743 C00489
66 58 Glyceric acid Glycerate (2,3- 17.68 740.0 113 497 PB 40 DI water 10 HMDB00139 439194 C00258
Dihydroxy
propionate)
67 35 Glycine Glycine 13.56 483.0 256 212 hF 200 DI water 100 HMDB00123 750 C00037
68 14 Glycolic acid Glycolate 9.35 484.0 285 213 PB 40 DI water 100 HMDB00115 757 C00160
69 125 Glycylproline Glycylproline 29.83 580.1 70 153 hF 40 0.1 M HCl 10 HMDB00721 79101
70 39a Hexanoylglycine Hexanoylglycine-1 14.07 155.1 99 71 hF 20 DI water 10 HMDB00701 99463
(cyclic)
(continued)
Table 1
(continued)
Diagnostic Stock
GC ions Internal L3 solution Metabolite database coding
The profiled
No. metabolites as the RT MM m/ c Stock
No. [12]a Traditional name HFBCF derivatives (min) product zq1 m/zq2 Standardb (μM)c solution c (mM) HMDB PubChem KEGG
71 78a Hippuric acid Hippurate-1 (cyclic, 19.86 161.0 105 161 hF 400 Ethanol 100 HMDB00714 464 C01586
60%)
72 106 Histamine Histamine 26.48 563.1 308 320 hF 40 0.1 M HCl 10 HMDB00870 774 C00388
73 114a Histidine Histidine-2 (NR) 27.9 789.0 307 362 hF 200 0.1 M HCl 100 HMDB00177 6274 C00135
74 100 Homocysteine Homocysteine 24.68 769.0 282 342 hF 40 MeCN 10 HMDB00742 778 C05330
(total)**
75 103 Homophenylalanine Homophenylalanine 25.98 587.1 91 283 0.1 M 100
(hF, I.S.) NaHCO3
76 104 Homovanillic acid Homovanillate 26.07 590.0 107 590 PB 20 0.1 M HCl 50 HMDB00118 1738 C05582
77 32 Hydroxyisocaproicacid 2-Hydroxyisocaproate 12.79 540.1 296 113 PB 20 DI water 10 HMDB00746 83697
78 118 Hydroxyphenyllactic 4-hydroxy- 28.54 816.0 572 345 PB 20 DI water 10 HMDB00755 9378 C03672
acid Phenyllactate
79 52 Hydroxypropionic acid 3-Hydroxypropionate 16.06 526.0 255 298 PB 100 DI water 10 HMDB00700 68152 C01013
(dimer)
80 113 Indolacetate Indolacetate 27.61 357.1 130 357 hF 20 MeCN 10
81 80 Isocitric acid Isocitrate 20.18 964.0 465 321 PB 100 DI water 100 HMDB00193 1198 C00311
82 50 Isoleucine Isoleucine 15.93 539.1 283 312 hF 40 0.1 M HCl 10 HMDB00172 6306 C00407
Diagnostic Stock
GC ions Internal L3 solution Metabolite database coding
The profiled
No. metabolites as the RT MM m/ c Stock
No. [12]a Traditional name HFBCF derivatives (min) product zq1 m/zq2 Standardb (μM)c solution c (mM) HMDB PubChem KEGG
83 59 Isovalerylglycine Isovalerylglycine 17.77 341.1 85 525 hF 40 DI water 10 HMDB00678 546304
84 6 Ketoleucine 2-Ketoisocaproate 7.43 312.1 85 57 PB 20 DI water 10 HMDB00695 70 C00233
85 126 Kynurenic acid Kynurenate 29.99 597.0 371 354 hF 40 NaHCO3 10 HMDB00715 3845 C01717
86 143 Kynurenine Kynurenine 35.07 842.1 146 372 hF 40 0.1 M HCl 10 HMDB00684 161166 C00328
87 13 Lactic acid Lactate 9.14 498.0 271 255 PB 100 DI water 100 HMDB00190 107689 C00186
88 49 Leucine Leucine 15.84 539.1 312 270 hF 40 0.1 M HCl 50 HMDB00687 6106 C00123
89 119a Lysine Lysine-2 (N,N-R) 28.79 780.1 310 256 hF 100 0.1 M HCl 100 HMDB00182 5962 C00047
90 51 Malic acid Malate 16.04 724.0 281 253 PB 40 DI water 10 HMDB31518 92824 C00497
91 7 Malonic acid Malonate 7.51 468.0 269 407 PB 40 Ethanol 10 HMDB00691 867 C00383
92 74 Mandelic acid Mandelate 19.45 560.0 289 333 PB 20 DI water 10 HMDB00703 439616 C01984
93 88 Methionine Methionine 21.65 557.0 61 357 hF 40 0.1 M HCl 10 HMDB00696 6137 C00073
94 117 Methioninesulfone Methionine sulfone 28.51 589.0 282 82 hF 40 0.1 M HCl 10
95 71 Methylcysteine S-Methylcysteine 19.13 543.0 61 300 hF 20 0.1 M HCl 10 HMDB02108 24417
96 33 Methylglutaric acid 3-Methylglutarate 12.93 510.1 311 282 PB 40 DI water 10 HMDB00752 12284
97 8 Methylmalonic acid Methylmalonate 7.54 482.0 283 438 PB 40 DI water 10 HMDB00202 487 C02170
98 18 Methylsuccinic acid Methylsuccinate 10.38 496.0 297 268 PB 40 DI water 50 HMDB01844 10349 C08645
99 66 N-Acetyl-asparticacid N-Acetylaspartate 18.38 539.0 270 312 hF 200 0.1 M HCl 50 HMDB00812 65065 C01042
100 28 Nicotinic acid Nicotinate 12.17 305.0 106 78 hF 40 0.1 M HCl 10 HMDB01488 938 C00253
(continued)
Table 1
(continued)
Diagnostic Stock
GC ions Internal L3 solution Metabolite database coding
The profiled
No. metabolites as the RT MM m/ c Stock
No. [12]a Traditional name HFBCF derivatives (min) product zq1 m/zq2 Standardb (μM)c solution c (mM) HMDB PubChem KEGG
101 109 Ornithine Ornithine 27.25 766.1 296 256 hF 40 0.1 M HCl 50 HMDB00214 6262 C00077
102 2 Oxalic acid Oxalate 5.38 454.0 113 183 PB 200 DI water 10 HMDB02329 971 C00209
103 42b Oxoglutaric acid 2-Ketoglutarate- 14.6 510.0 283 284 PB 200 DI water 100 HMDB00208 51 C00026
2(80%)
104 121 Palmitic acid Palmitate 28.91 438.2 255 438 PB 20 Ethanol 50 HMDB00220 985 C00249
105 31 Phenylacetic acid Phenylacetate 12.65 318.0 91 318 PB 20 Ethanol 10 HMDB00209 999 C07086
106 98 Phenylalanine Phenylalanine 23.39 573.1 91 330 hF 40 0.1 M HCl 100 HMDB00159 6140 C00079
107 84 Phenyllactic acid 3-Phenyllactate 20.92 574.0 330 131 PB 20 DI water 10 HMDB00779 3848 C01479
108 92 Phenylpyruvic acid Phenylpyruvate 22.84 572.0 118 329 PB 40 Ethanol 10 HMDB00205 997 C00166
109 61 Phthalic acid Phthalate 17.79 530.0 331 332 PB 20 DI water 10 HMDB02107 1017 C01606
110 56 Pimelic acid Pimelate 17.09 524.1 296 325 PB 20 Ethanol 10 HMDB00857 385 C02656
111 55 Proline Proline 16.84 523.0 296 297 hF 40 0.1 M HCl 10 HMDB00162 145742 C00148
112 142 Prolylhydroxyproline Prolylhydroxy 34.6 862.1 296 297 hF 100 0.1 M HCl 10 HMDB06695 11902892
proline
113 48 Propionylglycine Propionylglycine 15.55 313.1 57 56 hF 100 DI water 10 HMDB00783 98681
114 10 Propyl pentanoate 2-Propylvalerate 8.61 326.1 255 284 PB 40 DI water 10 HMDB40296 67328
115 69 Pyroglutamic acid Pyroglutamate 18.9 537.0 310 84 hF 200 DI water 100 HMDB00267 7405 C01879
116 73 Salicylic acid Salicylate 19.41 546.0 120 303 PB 20 DI water 10 HMDB01895 338 C00805
Diagnostic Stock
GC ions Internal L3 solution Metabolite database coding
The profiled
No. metabolites as the RT MM m/ c Stock
No. [12]a Traditional name HFBCF derivatives (min) product zq1 m/zq2 Standardb (μM)c solution c (mM) HMDB PubChem KEGG
117 120 Salicyluric acid 2-Hydroxy 28.82 603.0 120 403 hF 40 0.1 M HCl 10 HMDB00840 10253 C07588
hippurate
118 25 Sarcosine Sarcosine 12.07 497.0 270 226 hF 20 0.1 M HCl 10 HMDB00271 1088 C00213
119 95 Sebacic acid Sebacate 23.16 566.1 98 367 PB 40 Ethanol 10 HMDB00792 5192 C08277
120 77 Serine Serine 19.6 739.0 268 295 hF 100 DI water 100 HMDB00187 5951 C00065
121 134 Stearic acid Stearate 32.5 466.3 255 466 PB 20 Ethanol 50 HMDB00827 5281 C01530
122 72 Suberic acid Suberate 19.2 538.1 339 138 PB 20 MeCN 10 HMDB00893 10457 C08278
123 17 Succinic acid Succinate 10.32 482.0 283 55 PB 40 DI water 100 HMDB00254 1110 C00042
124 75 Thioproline Thioproline 19.46 541.0 314 287 hF 20 0.1 M HCl 10
125 57a Threonine Threonine-1(OH) 17.42 527.0 100 283 hF 100 0.1 M HCl 100 HMDB00167 6288 C00188
126 23 Tiglylglycine Tiglylglycine 11.27 339.1 83 55 hF 40 DI water 50 HMDB00959 6441567
127 139 Tryptamine Tryptamine 34.18 612.1 130 386 hF 20 0.1 M HCl 10 HMDB00303 1150 C00398
128 141 Tryptophan Tryptophan 34.44 612.1 130 131 hF 40 NaOH 100 HMDB00929 6305 C00078
129 127 Tyramine Tyramine 30.06 589.1 346 333 hF 20 Ethanol 10 HMDB00306 5610 C00483
130 129 Tyrosine Tyrosine 30.58 815.0 333 289 hF 40 0.1 M HCl 100 HMDB00158 6057 C00082
131 102 Urocanic acid Trans-urocanate 25.39 546.0 347 546 hF 20 DI water 10 HMDB34174 1549103
132 40 Valine Valine 14.1 525.1 298 283 hF 40 0.1 M HCl 100 HMDB00883 6287 C00183
133 128 Vanillactic acid 4-Hydroxy-3- 30.38 846.0 375 561 PB 20 DI water 10 HMDB00913 160637
methoxy
phenyllactate
134 122 Vanillylmandelic Vanillylmandelate 28.92 832.0 832 377 PB 20 0.1 M HCl 10 HMDB00291 736172 C05584
acid
MM = a molecular mass of each observed metabolite derivative; m/z = diagnostic ions in the EI spectrum of each metabolite; q1, q2 = diagnostic (quantitation and qualifier) fragment ions used for
the quantitative GC-SIM-EI-MS analysis
a
Numbering, traditional name, and metabolite product names according to the reference 12
b
Calibration against the internal standard; hF homophenylalanine, PB 4-phenylbutyric acid
c
Calibration level L3 (the observed average concentration in urine) [12]
174 Petr Hušek et al.
3 Methods
Table 2
GC-EI-SIM-MS operating conditions
3.4 GC-MS Analysis 1. The instrument GC-MS conditions are summarized in Table 2.
2. First, analyze the standard mixtures to check the separa-
tion performance, retention times, the analyte peak shape, and
GC-MS Metabolomic Profiling of Protic Metabolites 177
3.5 Data Analysis 1. Peak area for quantifier and qualifier ion of each metabolite is
integrated. Their ratio is calculated to test for potential
interferences.
2. The peak area of each quantifier is normalized by the peak area
of the corresponding internal standard: amino acids and bio-
genic amines against homophenylalanine, compound No. 75;
organic acids against 4-phenylbutyric acid, compound No. 37,
Table 1 [12].
3. Use appropriate vendor data processing software for data cali-
bration and metabolite quantification.
4. Check metabolite responses in the QC samples measured regu-
larly throughout the whole sample set. If the analyte’s relative
response to the IS fluctuates with RSD >30%, then even a semi-
quantitative measurement of such metabolite is difficult, and it
should be excluded from the metabolomic study (see Note 10).
5. Once the metabolite levels have been determined and met, pre-
defined acceptance criteria normalize appropriately the mea-
sured metabolite concentrations relative to creatinine. For urine
recalculate the metabolite levels to creatinine or other suitable
reference factors. Export the analytical data matrix into a
Microsoft Excel® spreadsheet or other formats suitable for fur-
ther chemometric analysis.
6. Use an appropriate statistical software to recognize differences
among the studied metabolite sample sets. The calculation of
178 Petr Hušek et al.
4 Notes
Acknowledgments
Abstract
Capillary electrophoresis (CE) is well suited for the profiling of highly polar and charged metabolites as
compounds are separated on the basis of their charge-to-size ratio. The protocol presented here is based
on using a recently developed sheathless interfacing design, i.e., a porous tip interface, for coupling CE to
electrospray ionization mass spectrometry (MS). It is demonstrated that sheathless CE-MS employing a
bare fused-silica capillary at low-pH separation conditions can be used for the profiling of both cationic
and anionic metabolites by only switching the MS detection and electrophoretic separation voltage polar-
ity. The proposed sheathless CE-MS protocol allows efficient and sensitive profiles to be obtained for a
broad array of charged metabolites, including amino acids, organic acids, nucleotides, and sugar phos-
phates, in various biological samples, such as urine and extracts of the glioblastoma cell line.
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_12, © Springer Science+Business Media, LLC, part of Springer Nature 2018
183
184 Rawi Ramautar
2 Materials
2.1 Solutions 1. Background electrolyte (BGE) solution: 10% (v/v) acetic acid,
and Samples pH 2.2. Add 9.0 mL of water into a 10 mL glass vial, and add
for Analysis 1.0 mL of acetic acid to the water in a fume hood. Mix the solu-
tion thoroughly using a vortex. Store at 4 °C.
2. Metabolite standard mixture: dissolve 50 μL of a 50 μM cation
standard mixture containing the basic twenty l-amino acids
into 50 μL of water, and mix the solution thoroughly (see Note
1). Store at −80 °C when not in use. Dissolve 50 μL of a 50 μM
anion standard mixture containing 17 anionic metabolites into
50 μL of water, and mix the solution thoroughly (see Note 1).
Store at −80 °C when not in use. The anionic metabolites
include (1) 2-naphthol-3,6-disulfonic acid; (2) d(+)2-
phosphoglyceric acid; (3) d-ribose-5-phosphate; (4) d-glucose-
1-phosphate; (5) d-glucose-6-phosphate; (6)
d-fructose-6-phosphate; (7) inosine 5′-monophosphate; (8)
guanosine 3′,5′-cyclic monophosphate; (9) guanosine 5′-mono-
phosphate; (10) citric acid; (11) trimesic acid; (12) isocitric
acid; (13) gluconic acid; (14) adenosine 3′,5′-cyclic monophos-
phate; (15) 2-hydroxybutyric acid; (16) b-diphosphopyridine
nucleotide (NAD+); and (17) 3-hydroxybutyric acid.
2.2 Analytical 1. The protocol reported here can only be performed with a com-
Equipment mercially available sheathless CE equipment, also known as
CESI 8000 (Sciex, A98089). Dependent on the type of MS
instrument, a dedicated nanospray source is required for
hyphenating sheathless CE to MS, information which can be
obtained from the vendor.
2. For the electrophoretic separations, commercially available
fused-silica capillaries (dimensions, 30 μm ID × 90 cm total
length) are used (Sciex, B07367).
3 Methods
The protocol described here for the use of sheathless CE-MS for
metabolic profiling studies is for laboratory use only. Prior to using
this protocol, consult all relevant material safety data sheets
(MSDS). Please use all appropriate laboratory safety procedures,
including safety glasses, lab coat, and gloves, when performing the
experiments described in this protocol.
3.1 Setting 1. Place a new bare fused-silica cartridge with a porous tip emitter
Up the CE System (30 μm ID × 90 cm total length) in the CE instrument.
2. Check for flow of liquid through the capillary by performing a
forward rinse at 50 psi for 15 min using 100% methanol (see
Note 2). Carry out also a rinse in the opposite direction at
186 Rawi Ramautar
50 psi for 5 min using 100% methanol to check the flow of liq-
uid through the conductive capillary (see Note 3).
3. Rinse the separation capillary with water at 50 psi for 10 min by
keeping the porous tip section, that is, the sprayer tip, in a
50 mL Falcon tube containing 5 mL of water.
4. Rinse the separation capillary with 0.1 M NaOH at 50 psi for
10 min, then by water at 50 psi for 10 min, and finally with
BGE at 50 psi for 10 min.
3.2 Coupling 1. Remove the sprayer tip of the fused-silica cartridge from the
Sheathless CE to MS water tube, and install it in the nanospray source adapter for
coupling to the MS instrument (see Note 4). The ESI voltage is
set to 0 during this step.
2. Ensure that the height of the BGE vials in the CE instrument
matches the height of the sprayer tip.
3. Check for flow of liquid through the conductive capillary by
rinsing with BGE at 50 psi for 5 min (see Note 5).
4. Rinse the separation capillary with BGE at 50 psi for 10 min in
the forward direction (see Note 2).
5. Position the porous tip emitter at the entrance of the MS inlet
at a distance of circa 2–3 mm. Apply a voltage of 30 kV using a
ramp time of 1 min, and start acquiring MS data in the m/z
range from 65 to 1000 m/z for metabolic profiling studies
using first an ESI voltage of 0 (see Note 6).
6. Set the ESI voltage to 1000 V while continue recording data.
Increase the ESI voltage with increments of 100 V until a con-
stant background signal is observed.
7. Optimize the porous tip emitter position with respect to the
center of the MS inlet by moving it in the x, y, or z-direction in
order to see which position provides the maximal and most sta-
ble MS signal (see Note 7).
8. After determining the optimal ESI voltage, set the ESI voltage
to 0, and decrease the CE voltage from 30 kV to 1 kV using a
ramp time of 5 min (see Note 8).
9. Create on the basis of the optimized parameters a CE-MS
method for the analysis of metabolite standards and biological
samples.
3.4 Preparation 1. Collect human urine samples of healthy subjects, pool the sam-
of Human Urine ples, and store at −80 °C prior to usage.
Samples 2. Prior to sheathless CE-MS analysis, mix the pooled urine sam-
ple with the BGE (1:1, v/v), and centrifuge for 10 min at 4 °C
and 16,100 × g.
Fig. 1 Multiple extracted ion electropherograms obtained for the analysis of anionic metabolite standards
metabolite (25 μM) with sheathless CE-MS in negative ion mode using a sheathless porous tip sprayer. Peaks:
(1) 2-naphthol-3,6-disulfonic acid; (2) d(+)2-phosphoglyceric acid; (3) d-ribose-5-phosphate; (4) d-glucose-1-
phosphate; (5) d-glucose-6-phosphate; (6) d-fructose-6-phosphate; (7) inosine 5′-monophosphate; (8) guano-
sine 3′,5′-cyclic monophosphate; (9) guanosine 5′-monophosphate; (10) citric acid; (11) trimesic acid; (12)
isocitric acid; (13) gluconic acid; (14) adenosine 3′,5′-cyclic monophosphate; (15) 2-Hydroxybutyric acid; (16)
b-Diphosphopyridine nucleotide (NAD+); (17) 3-Hydroxybutyric acid. Experimental conditions: BGE, 10% acetic
acid (pH 2.2); separation voltage, −30 kV (+0.5 psi applied at the inlet of the CE capillary); sample injection,
2.0 psi for 60 s (Reproduced from ref. 13 with permission from the authors)
Fig. 2 Extracted ion electropherogram obtained for the analysis of isoleucine and leucine (25 μM) with sheath-
less CE-MS in positive ion mode. Experimental conditions: BGE, 10% acetic acid (pH 2.2); separation voltage,
+30 kV; sample injection, 2.0 psi for 60 s
Fig. 3 Metabolic profile (total ion electropherogram) obtained for an extract of a glioblastoma cell line (cell
density ~20 cells/nL) with sheathless CE-MS in positive ion mode. Experimental conditions: BGE, 10% acetic
acid (pH 2.2); separation voltage, +30 kV; sample injection, 2.0 psi for 60 s (Reproduced from ref. 13 with
permission from the authors)
190 Rawi Ramautar
4 Notes
Acknowledgment
References
1. Ramautar R, Berger R, van der Greef J et al ery and clinical application. Proteomics Clin
(2013) Human metabolomics: strategies to Appl 9(3–4):322–334
understand biology. Curr Opin Chem Biol 7. Soga T, Ueno Y, Naraoka H et al (2002)
17(5):841–846 Simultaneous determination of anionic inter-
2. Kuehnbaum NL, Britz-McKibbin P (2013) mediates for Bacillus subtilis metabolic path-
New advances in separation science for metab- ways by capillary electrophoresis electrospray
olomics: resolving chemical diversity in a post- ionization mass spectrometry. Anal Chem
genomic era. Chem Rev 113(4):2437–2468 74(10):2233–2239
3. Theodoridis GA, Gika HG, Want EJ et al 8. Soga T, Ohashi Y, Ueno Y et al (2003)
(2012) Liquid chromatography-mass spec- Quantitative metabolome analysis using capil-
trometry based global metabolite profiling: a lary electrophoresis mass spectrometry.
review. Anal Chim Acta 711:7–16 J Proteome Res 2(5):488–494
4. Ramautar R (2016) CE-MS in metabolomics: 9. Maxwell EJ, Chen DD (2008) Twenty years
status quo and the way forward. Bioanalysis of interface development for capillary
8(5):371–374 electrophoresis-electrospray ionization-mass
5. Pejchinovski M, Hrnjez D, Ramirez-Torres A spectrometry. Anal Chim Acta
et al (2015) Capillary zone electrophoresis on- 627(1):25–33
line coupled to mass spectrometry: a perspec- 10. Bonvin G, Schappler J, Rudaz S (2012)
tive application for clinical proteomics. Capillary electrophoresis-electrospray
Proteomics Clin Appl 9(5–6):453–468 ionization-
mass spectrometry interfaces: fun-
6. Pontillo C, Filip S, Borras DM et al (2015) damental concepts and technical develop-
CE-MS-based proteomics in biomarker discov- ments. J Chromatogr A 1267:17–31
192 Rawi Ramautar
11. Bonvin G, Veuthey JL, Rudaz S et al (2012) 14. Hirayama A, Tomita M, Soga T (2012) Sheathless
Evaluation of a sheathless nanospray interface capillary electrophoresis-mass spectrometry with a
based on a porous tip sprayer for CE-ESI-MS high-sensitivity porous sprayer for cationic metab-
coupling. Electrophoresis 33(4):552–562 olome analysis. Analyst 137(21):5026–5033
12. Ramautar R, Busnel JM, Deelder AM et al 15. Moini M (2007) Simplifying CE-MS opera-
(2012) Enhancing the coverage of the urinary tion. 2. Interfacing low-flow separation tech-
metabolome by sheathless capillary niques to mass spectrometry using a porous
electrophoresis-mass spectrometry. Anal Chem tip. Anal Chem 79(11):4241–4246
84(2):885–892 16. Zhang W, Gulersonmez MC, Hankemeier T
13. Gulersonmez MC, Lock S, Hankemeier T et al et al (2016) Sheathless capillary electrophoresis-
(2016) Sheathless capillary electrophoresis- mass spectrometry for metabolic profiling of
mass spectrometry for anionic metabolic profil- biological samples. J Vis Exp (116).
ing. Electrophoresis 37(7–8):1007–1014 doi:https://doi.org/10.3791/54535
Part III
Plant/Food Applications
Chapter 13
Abstract
Metabolomics is the area of research, which strives to obtain complete metabolic fingerprints, to detect
differences between them, and to provide hypothesis to explain those differences [1]. But obtaining com-
plete metabolic fingerprints is not an easy task. Metabolite extraction is a key step during this process, and
much research has been devoted to finding the best solvent mixture to extract as much metabolites as
possible.
Here a procedure is described for analysis of both polar and apolar metabolites using a two-phase
extraction system. D2O and CDCl3 are the solvents of choice, and their major advantage is that, for the
identification of the compounds, standard databases can be used because D2O and CDCl3 are the solvents
most commonly used for pure compound NMR spectra. The procedure enables the absolute quantifica-
tion of components via the addition of suitable internal standards. The extracts are also suitable for further
analysis with other systems like LC-MS or GC-MS.
Key words Two-phase extraction, NMR, Metabolic fingerprints, Plants, Identification, Quantification
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_13, © Springer Science+Business Media, LLC, part of Springer Nature 2018
195
196 Jan Schripsema and Denise Dagnino
TMSP
D2O phase
T T
T
PPM 9.0 8.0 7.0 6.0 5.0 4.0 3.0 2.0 1.0 0.0
C C
CDCI3 phase
P C
P P
PPM
9.0 8.0 7.0 6.0 5.0 4.0 3.0 2.0 1.0 0.0
Fig. 1 1H NMR spectra from the (a) D2O and (b) CDCl3 extracts of coffee powder obtained with the two-phase
extraction. In the CDCl3 phase, signals from caffeine (marked with C) and pyridine (marked with P) are indi-
cated. In the D2O phase signals from TMSP and trigonelline (marked with T) are indicated
2 Materials
used and for the organic phase pyridine. The TMSP spectrum has
only one signal at 0.00 ppm making it suitable for both quantifica-
tion and as a reference for the chemical shift.
For the organic phase pyridine is suggested since it provides
some advantages. The spectrum of pyridine contains three signals
of which two (at 8.61 ppm and 7.69 ppm) are clearly separated
from other signals. The third signal (7.29 ppm) is close to the
residual solvent signal and is more difficult to integrate. TMS,
often used as chemical shift reference in chloroform, is not advised
for quantification purposes, because it is more volatile and its signal
is quite narrow. Furthermore its relaxation time is higher than
most other compounds [9].
In metabolomics experiments, many signals are not identified,
but the peak areas should be corrected to permit quantitative com-
parison between samples. First of all instead of the areas, the rela-
tive areas in relation to the area of the IS are used (area/area IS
signal). This relative area should then be corrected for eventual
deviations of the actual quantities of the IS added to the tube, the
quantity of plant material extracted, and the quantity of solvent
used to extract the plant material and of the liquid transferred to
the NMR tube. This leads to the following multiplication factor
(MF):
MF = ( Actual Weight IS sol. / Weight 100 …lIS sol.)
(1000 mg of plant material / actual Quant.plant material )
( Quant.extraction solvent / Quant.extraction solvent transferred ) .
This factor is the same for all signals in the NMR spectrum of
a sample. In this way direct comparison between samples is
possible.
If the compound of interest has well-resolved signals which
can be integrated, the absolute quantity of the compound can be
calculated using the following formula:
Quant.IS in 100 …lIS sol. (mg)
( area compound signal / area IS siggnal )
MF ( no.Hs IS signal / no.Hs compound signal )
( mol.weight compound / mol.weight IS) .
This formula provides the absolute quantity per gram of plant
material. These calculations are illustrated in Table 1 for specific
signals in the spectra shown in Fig. 1.
Table 1
Calculations with the NMR data shown in Fig. 1. For each spectrum the calculation of the absolute
quantity of a specific compound is illustrated
3 Methods
3.1 Extraction The extraction described here has been used for dried plant mate-
and Analysis rial (freeze dried) and also for the analysis of dried bacteria and
food stuffs such as teas, coffee, butter, and cheese. The procedure
200 Jan Schripsema and Denise Dagnino
Fig. 2 Schematic representation of the two-phase extraction for comprehensive analysis of the plant metabo-
lome by NMR
4 Notes
References
1. Schripsema J, Dagnino D (2015) Metabolomics. 5. Heyman HM, Meyer JJM (2012) NMR-based
In: Hostettmann K, Stuppner H, Marston A, metabolomics as a quality control tool for
Chen S (eds) Handbook of chemical and bio- herbal products. S Afr J Bot 82:21–32
logical plant analytical methods, 1st edn. Wiley, 6. Kim HK, Verpoorte R (2010) Sample prepara-
New York tion for plant metabolomics. Phytochem Anal
2. Schripsema J (2010) Application of NMR in 21:4–13
plant metabolomics: techniques, problems and 7. Beltran A, Suarez M, Rodríguez MA et al
prospects. Phytochem Anal 21:14–21 (2012) Assessment of compatibility between
3. Deda O, Gika HG, Wilson IA, Theodoridis GA extraction methods for NMR and LC/MS-based
(2015) An overview of fecal preparation for metabolomics. Anal Chem 84:5838−5844
global metabolic profiling. J Pharm Biomed 8. Kim HK, Choi YH, Verpoorte R (2010) NMR-
Anal 113:137–150 based metabolomic analysis of plants. Nat
4. Kim H-S, Park SJ, Hyun S-H et al (2011) Protoc 5:536–549
Biochemical monitoring of black raspberry 9. Schripsema J (2008) Comprehensive analysis of
(Rubus coreanus Miquel) fruits according to polar and apolar constituents of butter and mar-
maturation stage by 1H NMR using multi- garine by Nuclear Magnetic Resonance, reflect-
ple solvent systems. Food Res Int 44: ing quality and production processes. J Agric
1977–1987 Food Chem 56:2547–2552
Chapter 14
Abstract
NMR spectroscopy has become an indispensable tool for the metabolic profiling of foods and food prod-
ucts. In the present protocol, we report an analytical approach based on liquid-state NMR for the deter-
mination of polar and nonpolar metabolites in some common liquid (wine, spirits, juice) and solid (cheese,
coffee, honey) foods. Although the diversity of foods precludes the use of a single protocol, with small
modifications, the proposed methodologies can be adapted to a broader range of foodstuffs.
Key words NMR spectroscopy, Metabolite profiling, Food analysis, Authentication, Quality control
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_14, © Springer Science+Business Media, LLC, part of Springer Nature 2018
203
204 Evangelia Ralli et al.
2 Materials
Fig. 1 1H NMR spectra of coffee obtained by applying method in Subheading 3.2: lipid profile in CDCl3 (top) and
polar metabolite profile in D2O (bottom), both spectra obtained at a proton frequency of 500.13 MHz
2.3 Buffer Oxalate buffer (pH = 4): transfer 0.0595 g of oxalic acid and
Preparation 0.1795 g of sodium oxalate in a 20 mL volumetric flask, dissolve in
D2O (99.9 atom % D), and leave in ultrasound sonicator for
15 min.
Phosphate buffer (pH = 7.4): prepare phosphate buffer
(pH 7.4) by weighing 0.721 g Na2HPO4, 0.131 g NaH2PO4,
1 mM TSP (0.025 g), and 3 mM NaN3 (0.542 g) into a 25 mL
volumetric flask. Add 5 mL of D2O and fill up to 25 mL with
water. Shake thoroughly, and leave in a sonicator at 40 °C, inter-
spersed by shaking the flask, until the salts are dissolved.
3 Methods
3.1 Direct Analysis 1. Store 2 mL of sample in a 4 mL screw cap vial at −18 °C for
of Liquid or 12 h.
Water-Soluble Foods 2. Lyophilize for at least 12 h.
3.1.1 Analysis of Wine 3. Add 400 μL D2O containing 0.05% TMSP and 200 μL oxalate
buffer.
4. Centrifuge in an Eppendorf vial at 13, 1 48 × g for 10 min.
5. Transfer the supernatant into a 5 mm NMR tube.
6. Run NMR experiment Protocol A.
3.2.2 Polar Metabolite 6. Weigh 3× 0.30 g of ground sample into three Eppendorf vials
Extraction (total 0.90 g).
7. Add 1 mL of ultrapure water in each vial by a variable volume
pipette 100–1000 μL.
8. Seal vials with laboratory film, place into sonicator bath for
30 min, and centrifuge at 13,148 × g for 10 min (see Note 3).
9. Carefully remove the aqueous phase via pipette, and transfer
into a tarred 50 mL glass flask.
10. Re-extract the remaining pellet two more times (repeat steps
7–9).
11. After the extraction in triplicate, store the three Eppendorf
vials at −18 °C (to be used for the extraction of lipids in step
17) (see Note 4).
12. Freeze at −18 °C the glass flask with collected extracts and
freeze-dry for 16 h.
13. After freeze-drying, weigh the precipitates, and transfer
approx. half of the precipitate into a 4 mL screw cap glass vial.
14. Add 700 μL D2O containing 0.05% TMSP via a variable vol-
ume pipette in the glass vial, and place into ultrasound sonica-
tor bath for 30 min (see Note 5).
15. Filter the polar extracts through glass wool tightly packed into
a Pasteur pipette, directly into a 5 mm NMR tube.
16. Run NMR experiment Protocol B.
208 Evangelia Ralli et al.
3.2.3 Nonpolar
Metabolite Extraction 17. Add 1 mL chloroform in each Eppendorf vial obtained in step
11.
18. Seal the three Eppendorfs with laboratory film, place into
ultrasound bath for 30 min, and centrifuge at 10,000 rpm
(6,708 × g) for 10 min.
19. Carefully remove the liquid phase and transfer into a glass flask
of 50 mL.
20. Re-extract the remaining pellet two more times with chloro-
form (repeat steps 17–19).
21. Evaporate the chloroform from the glass flask in a rotary
evaporator.
22. Add 700 μL CDCl3 containing 0.03% TMS (by the use of vari-
able volume pipette 100–1000 μL) in the glass flask with the
dried extracts, and place flask into ultrasound bath for 5 min.
23. Filter extracts carefully through glass wool directly into a
5 mm NMR tube.
24. Run NMR experiment Protocol C.
3.3 NMR Set the probe temperature to 298 K, and wait (5–10 min) until the
Spectroscopy sample temperature is equilibrated. Lock, tune, and shim the sam-
Experimental ple according to standard NMR spectrometer procedures (see Note
Protocols 6).
3.3.1 Protocol A. 1H NMR
Spectroscopy WET 1. Obtain a 1H NMR spectrum (Bruker, zg30); integrate and save
Experiment the spectral regions containing the water and ethanol peaks to
be suppressed per automation program directions (see Note 7).
2. Load a standard WET solvent multisuppression (Bruker/WET)
pulse program with default spectrometer parameters.
3. Record a WET multisuppressed 1H NMR spectrum with param-
eters SW = 20 ppm, NS = 256 scans, DS = 8 dummy scans,
AQ = 3.3 s, and D1 = 1 s.
4. Perform Fourier transformation, phase correction, and baseline
correction according to standard NMR spectrometer (or pro-
cessing software) procedures.
3.3.2 Protocol B. 1H NMR 1. Obtain a 1H NMR spectrum (Bruker, zg30), and record the
Spectroscopy Water exact frequency of the residual water proton peak.
Presaturation Experiment
2. Load a standard solvent presaturation (Bruker, zgpr) pulse pro-
gram with default spectrometer parameters, and set the fre-
quency of the residual water signal exactly on resonance.
3. Record a water-suppressed 1H NMR spectrum with parameters
SW = 12 ppm, NS = 256 scans, DS = 4 dummy scans,
AQ = 5.45 s, and D1 = 1 s.
NMR Spectroscopy Protocols for Food Metabolomics Applications 209
3.3.3 Protocol C. 1H NMR 1. Load a standard proton NMR experiment (Bruker, zg30).
Spectroscopy Standard 2. Record a 1H NMR spectrum with parameters SW = 12 ppm,
Experiment TD = 64 K, NS = 256 scans, DS = 4 dummy scans, AQ = 3.3 s,
and D1 = 1 s.
3. Perform Fourier transformation, phase correction, and baseline
correction according to standard NMR spectrometer (or pro-
cessing software) procedures.
3.4 NMR Data The use of an internal standard in the form of TMSP allows the
Analysis quantitative determination of polar metabolites in the food sam-
ples from the integration of proton signals in the 1H NMR spectra
obtained. The 1H NMR spectra of the nonpolar fraction of solid
3.4.1 Metabolite Profiling foods (such as cheese, coffee, cocoa) can be used to quantify the
fatty acid profile of the lipid fraction of these foods. In either case,
identification of metabolites can be achieved through spiking
experiments (when the metabolite is expected or suspected) and
verified through 2D NMR spectroscopy. Other means of metabo-
lite identification include the use of publicly available NMR spec-
tral databases, such as FoodDB, HMDB, BMRB, etc. Multivariate
statistical analysis methods can be applied directly on the quantita-
tive NMR metabolite profiles, in order to develop models for
studying food authentication, quality control, cultivar, pedocli-
matic effects, etc.
3.4.2 Metabolite For metabolite fingerprinting applications, the whole NMR spec-
Fingerprinting trum is used for statistical analysis and development of multivariate
metabolomic models. The first step is to convert the NMR spectra
into ASCII files and/or use suitable software (AMIX) to segment
the NMR spectra into buckets, for variable reduction purposes.
The size (width) of the buckets is user-defined, usually between
0.005 and 0.01 ppm for 1H NMR data, and the spectra are nor-
malized to the total sum of integrals of all buckets. Regions of the
spectra that do not contain any signal or contain solvent signal
(e.g., water or ethanol) can be omitted from the bucketing proce-
dure. If the NMR spectra need alignment, due to pH, tempera-
ture, or other sample variations, this should be performed using
specialized software, before the bucketing procedure [19].
In the data matrix obtained, the different food samples make
up the rows, while the buckets make up the matrix columns,
respectively. The dataset can be subjected to multivariate statistical
analysis using a variety of commercial and academic/public soft-
ware packages, including STATISTICA, SIMCA, R, etc. Analysis
of variance (ANOVA) and unsupervised principal component
210 Evangelia Ralli et al.
4 Notes
Abstract
The instrumental characterization of volatile organic compounds (VOCs) is essential to have a precise,
reliable, and reproducible estimation of food aroma and, therefore, of the overall product quality. In this
report, we introduce four analytical approaches based on PTR-MS (proton transfer reaction-mass spec-
trometry) technology suitable to fully investigate the complexity of apple aroma. In our opinion, these
proposed methodologies can be applied, with slight modification, to every kind of fruit for destructive and
nondestructive rapid VOC fingerprinting.
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_15, © Springer Science+Business Media, LLC, part of Springer Nature 2018
213
214 Brian Farneti
1.3 Destructive The correlation between VOC emitted by an intact fruit and its
Analysis of Fresh Fruit internal content is still commonly accepted. Indeed, most of the
Tissue investigations carried out on VOC composition in fruit, and related
216 Brian Farneti
1.4 Dynamic The flavor of a product is not a stable trait, but it can rapidly and
Analysis by In Vitro drastically change during time, for instance, during fruit consump-
Mastication tion. Differences in VOC release behaviors may influence the
human aroma perception during food consumption since VOCs
are released from the matrix and then transported to mouth and
nose receptors.
In order to describe the release kinetics of VOCs while the
food matrix is being chewed, we developed an analytical system
based on an artificial chewing device coupled with the PTR-
ToF-MS. This system allowed a precise dynamic VOC fingerprint-
ing while the food is processed.
1.5 Automated The high biological variability between samples is one of the main
Analysis of Frozen risks that has to be considered during the design of an experiment
Fruit Tissue focused on VOC assessment. Therefore, a high number of biologi-
cal replicates are fundamental for a statistically correct experimen-
tal design. The possibility to couple a PTR-ToF-MS instrument
with a multipurpose sampler allowed the analysis of more than 200
samples a day with high reproducibility and with reduced lab labor.
The limiting factor of this methodology is, however, the restricted
volume of the vials that have to be used (20 ml). This restriction
can be exceeded by using powdered frozen tissue of the fruit (i.e.,
cortex or peel).
2 Materials
2.1 Nondestructive 1. Sealed glass jars with a volume of 1000 ml. Each jar lid needs
Analysis of Intact Fruit two entries (one inlet and one outlet) with the possibility to be
tightly closed.
2. A laboratory water bath.
3. An analytical balance.
4. A zero air producer.
5. PTR-ToF-MS 8000 instrument (Ionicon Analytik GmbH,
Innsbruck, Austria).
The PTR-ToF-MS drift tube is set with the following condi-
tions: 110 °C drift-tube temperature, 2.25 mbar drift pressure, and
550 V drift voltage. This leads to an E/N ratio of about 140
Townsend, where E corresponds to the electric field strength and
N to the gas number density.
Direct Injection Analysis of Fruit VOCs by PTR-ToF-MS: The Apple Case Study 217
The sampling time per channel of ToF acquisition was 0.1 ns,
amounting to 350,000 channels for a mass spectrum ranging up to
m/z = 400. Every single spectrum is the sum of about 28,600
acquisitions lasting 35 μs each, resulting in a time resolution of 1 s.
2.2 Destructive 1. Sealed glass jars with a volume of 250 ml. Each jar lid needs two
Analysis of Fresh Fruit entries (one inlet and one outlet) with the possibility to be
Tissue tightly closed.
2. A laboratory water bath.
3. A cork borer (diameter of 1.70 cm).
4. Homemade six-blade knife (1 cm distance between blades).
5. A zero air generator.
6. PTR-ToF-MS 8000 instrument (Ionicon Analytik GmbH,
Innsbruck, Austria). The PTR-ToF-MS setting is the same
described in Subheading 2.1.
Fig. 1 Schematic representation of the chewing device: cylindrical glass cuvette of 800 ml sealed with a cap
and a notched plunger controlled manually. The heat map indicates the dynamic VOC fingerprinting of apple
fruit assessed by PTR-ToF-MS coupled with the artificial chewing device. Three examples of VOC release dur-
ing the dynamic analysis are shown (m/z 61.028, 81.07, 45.033). Each graph is divided by a line, at time 0, in
two phases, respectively, before and after the chewing moment
3 Methods
3.1 Nondestructive 1. A whole intact fruit, without any damages, is weighted and
Analysis of Intact Fruit placed into a 1000 ml glass jar and incubated for 30 min at
30 °C into the water bath (see Note 1).
2. After the incubation, the headspace of the samples is directly
connected to the PTR-ToF-MS instrument via a heated PEEK
tube (110 °C. 0.055″ diameter) and sampled at a flow rate of
40 standard cm3 per min (sccm). At the same time, the jar is
continuously flushed with zero air in order to avoid air contami-
nation and under pressure (see Notes 2–9).
3. Sampling measurement was performed over 60 cycles resulting
in an analysis time of 60 s/sample.
3.2 Destructive 1. A cylindrical portion of cortex tissue is sampled with the cork
Analysis of Fresh Fruit borer along the vertical lengthwise plane of the fruit, avoiding
Tissue the core portion with seeds.
2. From this cylinder, five identical disks with a diameter of
1.70 cm and 1 cm thick are cut with a homemade six-blade
knife. Adopting this strategy, we avoid any possible influence of
fruit size, and we have tissue samples of different fruit regions.
3. These five disks are placed into a 250 ml glass jar and incubated
for 30 min at 30 °C into the water bath (see Note 1).
Direct Injection Analysis of Fruit VOCs by PTR-ToF-MS: The Apple Case Study 219
3.5 Data Analysis The analysis of PTR-ToF-MS spectral data (Fig. 2) proceeded cor-
recting offline the count loss (due to the ion detection dead time)
through Poisson statistics, following the method reported by
Cappellin et al. [8], while the internal calibration was performed
according to the procedure described in Cappellin et al. [9] (see
Notes 10–20). Such approach allowed a mass accuracy higher
than 0.001 Th to be achieved sufficient for the sum formula deter-
mination in our case. Compound annotation was carried out com-
paring the spectral profile with fragmentation data of reference
standards. Noise reduction, baseline removal, and peak intensity
extraction were performed according to Cappellin et al. [9], using
modified Gaussians to fit the peak shapes. Absolute headspace
VOC concentrations, expressed in ppbv (parts per billion by vol-
ume), were calculated from peak intensities according to the for-
mula described by Lindinger et al. [2]. A constant reaction rate
coefficient of 2 × 10−9 cm3/s was used in the calculations, intro-
ducing a systematic error of up to 30% that can be accounted for if
the actual rate coefficient is known [10].
Fig. 2 Example of PTR-ToF-MS spectrum of apple headspace. The main figure represents a sample spectrum
in the mass region between 1 and 420 Th, while the second figure enlarges the region around a selected nomi-
nal mass peak (m/z 31)
Direct Injection Analysis of Fruit VOCs by PTR-ToF-MS: The Apple Case Study 221
4 Notes
References
Abstract
This chapter describes a protocol for the analysis of the metabolomic fingerprint of wine by liquid
chromatography-mass spectrometry. The straightforward, optimized sample preparation procedure is lim-
ited to a single-step dilution with water or acetonitrile. The separation of wine analytes is carried out by
two columns with orthogonal selectivity, including both reversed-phase (C18) and hydrophilic interaction
(HILIC) chromatography, while the detection is assured by a high-resolution quadrupole time-of-flight
mass spectrometer operating in negative and positive electrospray ionization mode, in order to obtain four
different chromatograms for each sample. This validated protocol, or parts of it, could be applied in several
oenological topic experimental designs, including wine quality and wine authenticity.
Key words Vitis vinifera, Grape, Food, Holistic, Metabolomics, Mass spectrometry, Liquid chroma-
tography, HILIC
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_16, © Springer Science+Business Media, LLC, part of Springer Nature 2018
225
226 Panagiotis Arapitsas and Fulvio Mattivi
Fig. 1 Typical BPI LC-MS chromatogram of the same wine sample analyzed in four different modes: (a) HILIC
UPLC-QTOF MS ESI–, (b) HILIC UPLC-QTOF MS ESI+, (c) reversed-phase UPLC-QTOF MS ESI+, and (d)
reversed-phase UPLC-QTOF MS ESI–. From these chromatograms, it is clear that each mode covers different
parts of the wine metabolome, although some metabolites would be registered from more than one platform
Fig. 2 (a) PCA plot of a reversed-phase LS-MS analysis, produced with the described protocol. The experimen-
tal design was based in 12 different wines produced by six grape varieties (five Pinot gris, three Grillo, one
Chardonnay, one Traminer, one Muller, and one Inzolia wines). For each wine eight different bottles were ana-
lyzed. The QC sample injections form a tight cluster in the center of the plot, indicating the quality of the data
set. Injections of the same wine biological replicates cluster together, indicating that this method could be used
for wine authenticity. (b) PCA plot of a HILIC-MS analysis, produced with the described protocol. Also here
injections belonging in the same wine clustered together. The numbers which tagged the QCs indicate the
order of the injection in the beginning of the sequence. In this experiment, eight injections were necessary to
equilibrate the LC-MS; that number is expected for a HILIC-MS untargeted experiment
2 Material
2.1 Analytical The LC system used is a Waters Acquity UPLC, which included a
Instrumentation quaternary solvent manager, a sample manager, and a column
manager. For the reversed-phase LC, the column used is a Waters
Acquity UPLC 1.8 μm 2.1 × 150 mm HSS T3 column and for the
normal phase LC/HILIC, a Waters Acquity BEH amide column
2.1 mm × 100 mm × 1.7 μm (HILIC type), equipped with an
Acquity UPLC BEH Amide 1.7 μm VanGuard pre-column. The
MS system is a Waters Synapt HRMS QTOF MS.
2.2 Solutions Mobile phase A: a 1 L volumetric flask is half filled with water, then
1 mL of formic acid is added, and finally the flask is filled to volume
with water.
Mobile phase B: a 1 L volumetric flask is half filled with metha-
nol, then 1 mL of formic acid is added, and finally the flask is filled
to volume with methanol.
A stock solution of HCOONH4 4 M is prepared in advance
and stored at 4 °C. In detail, 12.6 g of HCOONH4 is weighed in
a beaker, and about 50 mL of water is added. The solution is stirred
for 4 h and then transferred to a 100 mL volumetric flask and filled
to volume with water.
For the mobile phase C, 5 mL of the HCOONH4 4 M stock
solution is added to a 1 L volumetric flask containing 950 mL of
water, 1 mL of NH4OH is added, and then the flask is filled to
volume (HILIC-LC/MS ESI-). For the mobile phase D, a 1 L
volumetric flask is half filled with acetonitrile, then 1 mL of
NH4OH is added, and finally the flask is filled to volume with ace-
tonitrile (HILIC-LC/MS ESI–) (see Notes 2 and 3).
For the mobile phase E, 5 mL of the HCOONH4 4 M stock
solution is added to a 1 L volumetric flask containing 950 mL of
water, 1 mL of formic acid is added, and then the flask is filled to
volume (HILIC-LC/MS ESI+). For the mobile phase F, a 1 L
volumetric flask is half filled with acetonitrile, then 1 mL of formic
acid is added, and finally the flask is filled to volume with acetoni-
trile (HILIC-LC/MS ESI+)) (see Notes 2 and 3).
Seal wash solvent is a mixture of 500 mL of water and 500 mL
of methanol.
Weak needle wash for the reversed-phase LC (and strong nee-
dle wash for the normal phase/HILIC) is a mixture of 100 mL
methanol and 900 mL water. Strong needle wash for the reversed-
phase LC (and weak needle wash for the normal phase/HILIC) is
230 Panagiotis Arapitsas and Fulvio Mattivi
3 Methods
3.1 QTOF MS The MS data are collected using different runs in positive and neg-
Parameters ative ESI mode over a mass range of 50–2000 amu for RP-LC and
30–1000 for HILIC with scan duration of 0.3 s in centroid mode.
The instrument operated in W mode. The transfer collision energy
and trap collision energy are set at 6 and 4 V. The source parame-
ters are set as follows: capillary 3 kV for positive scan and 2.5 kV
for negative scan, sampling cone 25 V, extraction cone 3 V, source
temperature 150 °C, desolvation temperature 500 °C, desolvation
gas flow 1000 L/h, and nebulizer gas 50 L/h.
External calibration of the instrument is performed at the
beginning of each batch of analysis by direct infusion of a sodium
formate solution (10% formic acid/0.1 M NaOH/CH3CN at a
ratio of 1/1/8) by controlling the mass accuracy from 50 to
2000 m/z for RP-LC and from 30 to 1000 m/z for HILIC (less
than 5 ppm) and mass resolution (over 14,000 FWHM). Lock
mass calibration is applied using a solution of leucine enkephalin
(0.5 mg/L, m/z 556.2771 for ESI+ and 554.2620 for ESI- mode)
at 0.1 mL/min. LC and MS instruments of similar specifications
can be used.
3.2 LC Parameters The column used is held at 40 °C during the analysis, the injection
volume is 10 μL, the flow rate is 0.28 mL/min, and the samples
3.2.1 RP-LC Instrumental are kept at 4 °C throughout the analysis. The multistep linear gra-
Parameters dient used is as follows (mobile phases A and B): 0–1 min, 100%
mobile phase A isocratic; 1–3 min, 100–90% A; 3–18 min, 90–60%
A; 18–21 min, 60–0% A; 21–25.5 min, 0% A isocratic; 25.5–
25.6 min, 0–100% A; and 25.6–28 min 100% isocratic (see Note
3).
3.2.2 HILIC Instrumental
Parameters The column and pre-column are held at 50 °C during the analysis,
the injection volume is 10 μL, the flow rate is 500 μL/min, and the
samples are kept at 4 °C throughout the analysis. For the ESI- MS,
the mobile phases C and D are used, and the multistep gradient
elution is as follows: 0–1 min, 88% mobile phase C; 1–2 min,
88–80% of C; 2–10 min, 80–72% C; 10–11 min, 72–50% C;
11–13 min, 50% C; 13–13.1 min, 50–88% C; and 13.1–18 min,
88% C. For the ESI+ MS, the mobile phases E (instead of C) and
F (instead of D) are used, under the same multistep gradient (see
Note 3).
Wine Metabolome 231
3.3 Sample Once the sampling is completed and all the samples are in the labo-
Preparation ratory, wines should be codified according to a randomized
sequence, so the sample preparation and analysis can be completed
following this randomized sequence (see Note 4).
Wines are uncorked under nitrogen atmosphere (see Note 5),
and an aliquot is transferred into a 15 mL amber vial (filled to
capacity). Then, again under nitrogen atmosphere, a QC pooled
sample is prepared using 0.5 mL of each sample, and this is treated
in the same way as the study samples. The QC can be used to opti-
mize the optimum dilution for the LC-MS system used and the
number of the samples (see Notes 6 and 7). For a Waters Acquity
UPLC-Synapt QTOF MS system used for this protocol, the fol-
lowing sample preparation is optimized for experimental designs
including 60–200 commercial wines (see Notes 8 and 9).
10. Load the UPLC method and wait until the back pressure
equilibration (psi delta <20).
3.5 Sample The samples should be injected according to the randomized order
Sequence established before the sample preparation (see Subheading 3.3).
Then the analysis should be made according to the following
sequence (see Notes 3, 6–9, 11–15):
1. First run a blank sample injection.
2. Then run four QC injections for the reversed phase and ten QC
injections for the normal phase LC, in order to equilibrate the
column (Fig. 2).
3. Now you can start the injection of the real samples, but for
every six real sample injections, one QC sample should be
injected.
4. The sequence should end with one QC sample and finally one
blank sample injection.
3.6 Data Analysis Data analysis in metabolomics follows a specific but very wide
workflow [11, 12] that should include the following steps:
1. Data preprocessing, divided in deconvolution, peak picking, fil-
tering (optional), alignment, and bucketing (binning).
2. Data pretreatment, subdivided into normalization (optional),
centering (optional), scaling, managing missing values, and
managing outliers.
3. Data processing, employing the use of statistical tools such as
multivariate supervised and unsupervised analysis.
4. Data visualization, such as PCA plots (Fig. 2).
5. Data quality validation.
6. Marker detection, annotation, and interpretation.
The researcher can use various informatics tools (one or more)
to accomplish the above workflow, based in his/her experience/
knowledge and financial support. For example, XCMS online [12]
is a valid, free, and widely used tool. In this protocol, we propose
the use of the commercial and user-friendly software Progenesis QI
(Waters, Nonlinear Dynamics). Other useful open source
informatics tools are mzMine [13], MetaDB [11], MetAlign [14],
and MetaboAnalyst [15].
Between the objects of this protocol, the marker detection and
validation, metabolite annotation, or the hypothesis generation is
not included. For this reason, the data analysis will be concluded
with the data visualization (Fig. 2) which is important to validate
the quality of the data set produced by the instrumental analysis
(see Note 15).
Progenesis QI steps:
Wine Metabolome 233
4 Notes
3. Prepare all the necessary mobile phases 1 day before the analy-
sis (especially for HILIC), and load all the volume in the LC
bottles from the beginning. Avoid adding mobile phase dur-
ing the analysis.
4. It is important to collect, register, and keep track of the details
and the characteristics of all samples analyzed. MetaDB [11] is
a useful tool for this aim. Register all the meta-data informa-
tion of the samples according to the minimum reporting stan-
dards for plant biology context information in metabolomics
studies [16].
5. N2 atmosphere is important to avoid possible O2-driven
reactions.
6. If the QC sample is a pooled sample, prepared by mixing equal
aliquots of each individual sample of the sample set, can pro-
vide import help for (a) training, (b) method development/
adaptation, (c) column equilibration, (d) data quality control,
and (e) marker quality evaluation.
7. Surrogate QC samples can be used in long-term studies which
include more than one sequence. Standard mixes are not con-
sidered good QC solution in the metabolomics field.
8. For commercial light-bodied white wines, it is possible to per-
form 200 injections without losing sensitivity. For full-bodied
red wines, the maximum injection suggested for this sample
preparation/instrumental setup is around 80 injections.
9. In case of a large number of samples (over 200), consider the
possibility to divide the sample set in smaller subsets. In this
case instrumental and data analysis should be made separately,
and then compared for common markers. All sample subsets
should have equally divided groups, i.e., same number of con-
trol and treatment condition samples, and not one subset with
the control samples and one subset with all the treatment
samples.
10. Degas the water used for the wine dilution.
11. Clean the LC tubing and pumps with a solution of 25% meth-
anol, 25% acetonitrile, 25% isopropanol, and 25% water every
2 weeks for reversed phase and every 5 days for HILIC.
12. If standard mixes are included in the sequence, inject them (a)
in the beginning of the sequence after the blanks and before
the QCs and (b) in the end of the sequence after the last QC
and before the last blank injection.
13. If blanks or standard mixes are included during the injections
of the real samples, run a minimum of two QC injections
before continuing with the real sample injections.
Wine Metabolome 235
14. Monitor during the sequence (a) the LC back pressure, (b)
retention time of specific metabolites, (c) mass error for spe-
cific metabolites, and (d) the area for specific metabolites.
15. Use data visualization with unsupervised multivariate statisti-
cal tools, such as PCA plot, to control the data quality, a step
which should be frequently monitored during the sequence of
analysis, by examining the QC distribution/clustering in the
PCA plot and/or possible outliers in the PCA plot (Fig. 2). If
you have to reinject—or re-prepare and reinject—more
sample(s), it is better to include it/them in the end of the
sequence before the last QC.
References
1. Nicholson JK, Lindon JC (2008) Systems biol- 10. Mattivi F, Arapitsas P, Perenzoni D et al (2015)
ogy: metabonomics. Nature 455:1054–1056 Influence of storage conditions on the compo-
2. Gika HG, Theodoridis GA, Vrhovsek U et al sition of red wines - advances in wine research -
(2012) Quantitative profiling of polar primary ACS symposium series. In: ACS symposium
metabolites using hydrophilic interaction ultrahigh series. ACS Publications, Washington, DC,
performance liquid chromatography-tandem mass pp 29–49
spectrometry. J Chromatogr A 1259:121–127 11. Franceschi P, Mylonas R, Shahaf N et al (2014)
3. Theodoridis G, Gika H, Franceschi P et al MetaDB a data processing workflow in untar-
(2011) LC-MS based global metabolite profil- geted MS-based metabolomics experiments.
ing of grapes: solvent extraction protocol opti- Front Bioeng Biotechnol 2:72
misation. Metabolomics 8:175–185 12. Smith CA, Want EJ, O' Maille G et al (2006)
4. Theodoridis GA, Gika HG, Want EJ et al XCMS: processing mass spectrometry data for
(2012) Liquid chromatography–mass spec- metabolite profiling using nonlinear peak
trometry based global metabolite profiling: a alignment, matching, and identification. Anal
review. Anal Chim Acta 711:7–16 Chem 78(3):779–778
5. Naz S, Vallejo M, García A et al (2014) Method 13. Katajamaa M, Miettinen J, Oresic M (2006)
validation strategies involved in non-targeted MZmine: toolbox for processing and visual-
metabolomics. J Chromatogr A 1353:99–105 ization of mass spectrometry based molecular
6. Buscher JM, Czernik D, Ewald JC et al (2009) profile data. Bioinformatics 22:634–636
Cross-platform comparison of methods for 14. Lommen A (2009) MetAlign: interface-driven,
quantitative metabolomics of primary metabo- versatile metabolomics tool for hyphenated
lism. Anal Chem 81:2135–2143 full-scan mass spectrometry data preprocess-
7. Cajka T, Fiehn O (2016) Toward merging ing. Anal Chem 81:3079–3086
untargeted and targeted methods in mass 15. Xia J, Wishart DS et al (2016) Using MetaboAnalyst
spectrometry- based metabolomics and 3.0 for comprehensive metabolomics data analysis.
Lipidomics. Anal Chem 88:524–545 Curr Protoc Bioinformatics 55:14.10.1–14.10.91.
8. Arapitsas P, Speri G, Angeli A et al (2014) The https://doi.org/10.1002/cpbi.11
influence of storage on the “chemical age” of 16. Sumner LW, Amberg A, Barrett D et al
red wines. Metabolomics 10:816–832 (2007) Proposed minimum reporting stan-
9. Arapitsas P, Ugliano M, Perenzoni D et al dards for chemical analysis chemical analysis
(2016) Wine metabolomics reveals new sulfo- working group (CAWG) metabolomics stan-
nated products in bottled white wines, pro- dards initiative (MSI). Metabolomics
moted by small amounts of oxygen. 3:211–221
J Chromatogr A 1429:155–165
Part IV
Abstract
The use of tissue as a matrix to elucidate disease pathology or explore intervention comes with several
advantages. It allows investigation of the target alteration directly at the focal location and facilitates the
detection of molecules that could become elusive after secretion into biofluids. However, tissue metabolo-
mics/metabonomics comes with challenges not encountered in biofluid analyses. Furthermore, tissue
heterogeneity does not allow for tissue aliquoting. Here we describe a multiplatform, multi-method work-
flow which enables metabolic profiling analysis of tissue samples, while it can deliver enhanced metabo-
lome coverage. After applying a dual consecutive extraction (organic followed by aqueous), tissue extracts
are analyzed by reversed-phase (RP-) and hydrophilic interaction liquid chromatography (HILIC-) ultra-
performance liquid chromatography coupled to mass spectrometry (UPLC-MS) and nuclear magnetic
resonance (NMR) spectroscopy. This pipeline incorporates the required quality control features, enhances
versatility, allows provisional aliquoting of tissue extracts for future guided analyses, expands the range of
metabolites robustly detected, and supports data integration. It has been successfully employed for the
analysis of a wide range of tissue types.
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_17, © Springer Science+Business Media, LLC, part of Springer Nature 2018
239
240 Panagiotis A. Vorkas et al.
2 Materials
All solvents used should be LC-MS grade. All solvent mixtures are
described as volume parts per solvent. Efforts should be taken to
minimize the presence of contaminants in containers used (see
Note 1).
Tissue Multiplatform-Based Metabolomics/Metabonomics for Enhanced Metabolome… 241
2.2 Tissue Extraction 1. Bead beating: bead beating tubes (2 mL), filled with 1 mm zir-
conium beads. Zirconium beads should cover just above the
curved bottom of the tube (see Notes 3 and 4).
2. Bead beater (Bertin Technologies).
3. Solvent mixture for organic extraction: methanol/methyl tert-
butyl ether (MTBE) (1:3).
4. Solvent mixture for aqueous extraction: water/methanol
(MeOH) (1:1)
5. Centrifuge (e.g., Eppendorf).
6. Eppendorf tubes 1–2 mL (see Note 5).
7. Solvent mixture evaporation using vacuum concentrator work-
ing at 30 °C (2 h for organic and 2.5 h for aqueous extraction)
(see Note 6).
2.3.3 NMR Spectroscopy 1. Reconstitution buffer: for 1 L of buffer, mix 500 mL of high
purity water with 21.73 g Na2HPO4 (anhydrous), 2.63 g
NaH2PO4 (anhydrous), and 0.1 g sodium 3-trimethylsilyl-1-
[2,2,3,3,-d4] propionate (TSP) (0.01% m/v). Mix well and
then add 200 mL of D2O. Mix until salts are completely dis-
solved. Sonicate the solution or use magnetic spinner if neces-
sary. Adjust to pH 7.4 with NaOH crystals or HCl (high
Tissue Multiplatform-Based Metabolomics/Metabonomics for Enhanced Metabolome… 243
2.4.2 NMR Spectroscopy MATLAB (version 2014a) programming language is utilized for
importing and processing of NMR data, using in-house developed
scripts.
2.6 Pathway For pathway mapping the KEGG database [14] and Ingenuity
Mapping Pathway Analysis software (QIAGEN) is used.
3 Methods
Fig. 1 An overview of the tissue metabolomics/metabonomics multiplatform procedure. The procedure is com-
prised of seven basic steps: (1) tissue dissection, (2) metabolite extraction from the tissue sample (two con-
secutive extractions: organic, round 1, followed by aqueous, round 2), (3) data acquisition (NMR, HILIC-, and
RP-UPLC-MS), (4) data deconvolution and processing, (5) statistical analysis, (6) metabolite structure assign-
ment, and (7) data integration and biological interpretation. MeOH, Methanol; MTBE, methyl tert-butyl ether
246 Panagiotis A. Vorkas et al.
3.4 UPLC-MS 1. Add 200 μL of the lipid profiling reconstitution solvent mixture
Analysis and Data to the Eppendorf tubes of the extracts and blanks (see Note
Treatment 21).
2. Vortex vigorously for 1 min.
3.4.1 Reconstitution
3. Centrifuge for 20 min at 20,000 rcf at 4 °C.
Lipid Profiling
4. Decant the supernatant into an LC-MS total recovery vial.
5. Combine 50 μL of the reconstitute to produce the QC pooled
sample (see Notes 14 and 22).
Tissue Multiplatform-Based Metabolomics/Metabonomics for Enhanced Metabolome… 247
Fig. 2 Characteristic chromatograms/spectra from RP-UPLC-MS analysis of organic extracts (lipid profiling) in
(a) positive and (b) negative modes, HILIC-UPLC-MS analysis of aqueous extracts in (c) positive and (d) nega-
tive modes, and NMR spectra of aqueous extracts acquired using the standard sequence (gray line; prior to
baseline correction) and CPMG sequence (black line)
A range of compounds that are regularly detected in tissue samples, using the described UPLC-MS methods. Signals from these compounds can be used
to assess adduct formation, mass accuracy, chromatographic peak width/resolution, and retention time shifting
3.4.4 Data Normalization Normalize the data using total area or median fold change normal-
ization (see Note 30). There are cases where normalization might
not be necessary.
3.4.5 Statistical Analysis 1. Import data in SIMCA along with demographic data.
and Quality Control 2. Transform and scale the data. For UPLC-MS data a logarithmic
transformation followed by scaling is usually recommended.
The exploration of various transformation and scaling options
according to methodology, instrumentation, and acquired data
should be considered.
3. Use OPLS-DA to remove features attributed to the procedure
or contamination, by comparing samples against blanks, as pre-
viously described [7]. Typically, a cutoff of P(corr) > 0.5 should
be used in order to exclude the features attributed to
contaminants.
4. Use PCA to evaluate QC samples repeatability. Remove con-
tamination of the QC samples, if present, using PCA or
OPLS-DA (Fig. 3). If the latter method is used, a cutoff of
P(corr) > 0.8 is typically selected. Remove only features that are
exclusively present in the QC samples (see Note 31).
5. Use PCA for QC dilution feature assessment. Remove features
that do not respond to dilutions as previously described [7]. It
is recommended to only remove features that elute with the
solvent front, or correspond to lipid signals in areas of high
coelution of lipids of the same class in HILIC (these areas can
be found in previously described in-house databases of anno-
tated lipid species of the HILIC method) [7]. The QC dilution
series can also be retrospectively employed to further assess the
quality of the discovered putative markers of the studied disease
or intervention.
6. Proceed to further statistical analyses and group comparisons.
Fig. 3 The quality control (QC) samples may be containing additional contamination, as compared to samples,
due to differences between sample/QC preparation. A simple procedure can be followed to remove the fea-
tures associated with this contamination using multivariate statistics. (a) The PCA scores plot demonstrates a
contamination in the QC samples, which is not present in the individual samples. (b) The loadings plot can
assist in pinpointing the contaminating features. (c) Feature intensity per sample can help identify features that
are only present in QC samples and constitute a contaminant. (d) OPLS-DA scores plot. OPLS-DA can be
employed as an alternative method for identifying and removing contaminants. (e) The S-plot can be used to
visualize the features for exclusion. (f) The PCA scores plot after features corresponding to contaminants have
been removed, showing the QC samples clustering in the center of the individual samples
3.5 NMR Analysis Two aliquots (corresponding to 50 mg of tissue sample) are rec-
and Data Treatment ommended for the NMR acquisition:
3.5.1 Reconstitution
1. Add 700 μL of sodium phosphate buffer into the dried tissue
for Aqueous Extracts
extract.
2. Vortex for 1 min followed by sonication for 5 min.
3. Centrifuge for 20 min at 20,000 × g at 4 °C.
4. If two aliquots are used, transfer the supernatant from the first
tube to the second, and repeat steps 2 and 3.
5. Transfer 600 μL of the supernatant into an NMR tube.
6. Perform the same procedure for extraction blanks.
7. Load a small number of tubes with the buffer only, to test for
contamination.
3.5.3 Data Deconvolution The following steps are performed in MATLAB using in-house
and Processing developed scripts:
1. Perform phasing, baseline correction, and calibration using the
TSP signal.
2. Remove the TSP and water signals from the spectra.
3. Normalize if necessary. Normalize the data using total area or
median fold change normalization (see Note 30).
4. Align spectra if necessary.
Tissue Multiplatform-Based Metabolomics/Metabonomics for Enhanced Metabolome… 255
3.6 Biological For the processes of correlation network construction and pathway
Interpretation mapping, a table of metabolites that demonstrated statistically sig-
nificant alterations should be produced, along with statistical
power, fold change, and values of each metabolite per sample. It is
also recommended to assign to each metabolite its corresponding
KEGG ID to facilitate importing in pathway mapping software.
256 Panagiotis A. Vorkas et al.
3.6.1 Data Integration Several approaches can be employed for data integration depend-
ing on the biological question and techniques/methods used:
1. Combine two or more data matrices by using multiblock multi-
variate methods, such as O2PLS and OnPLS [19], where
extraction of predictive components can be achieved.
2. Perform correlation analysis (see Subheading 3.6.2) to obtain
associations between entities from the same or different experi-
ments/datasets. Collectively, these associations could lead to
inferences assisting in the biological interpretation of the dys-
regulation or intervention studied.
3. Use appropriate informatics tools (described in Subheading
3.6.3) to perform metabolite enrichment of altered metabolites
detected from the same or different experiments. Additionally,
metabolite/gene enrichment can be combined if such data are
available.
3.6.2 Correlation Construct correlation networks after calculating the Spearman cor-
Analysis relation coefficients. Correlation coefficients along with their cor-
responding entity pairs, which could be paired combinations of
metabolites, proteins, genes, demographics etc., can then be trans-
ferred to the Cytoscape software, where the correlation network
can be assembled. All presented correlations in the network should
have a p < 0.05 (after multiple testing correction), while the cutoff
of the correlation coefficient should be set based on the complexity
of the illustrated network. The correlation matrix can be demon-
strated either as a matrix table or as a heat map.
3.6.3 Pathway Mapping Import the list of metabolites/KEGG IDs in the KEGG mapper
and/or Ingenuity Pathways Analysis software to map the pathways
corresponding to the assigned discriminant metabolites. When
available, include data from additional biological levels, such as
protein and mRNA, in order to combine metabolite and gene
enrichment. This can result in higher confidence and statistical
power of results.
4 Notes
Acknowledgments
References
1. Lamour SD, Veselkov KA, Posma JM et al rotic plaques relate to stroke risk: an explor-
(2015) Metabolic, immune, and gut microbial atory study. Eur J Vasc Endovasc 52:5–10.
signals mount a systems response to Leishmania https://doi.org/10.1016/j.ejvs.2016.01.022
major infection. J Proteome Res 14:318–329. 12. Anwar MA, Adesina-Georgiadis KN, Spagou K
https://doi.org/10.1021/pr5008202 et al (2017) A comprehensive characterisation
2. Folch J, Lees M, Sloane Stanley GH (1957) A of the metabolic profile of varicose veins; impli-
simple method for the isolation and purifica- cations in elaborating plausible cellular path-
tion of total lipides from animal tissues. J Biol ways for disease pathogenesis. Sci Rep 7:2989.
Chem 226:497–509 https://doi.org/10.1038/
3. Bligh EG, Dyer WJ (1959) A rapid method of s41598-017-02529-y
total lipid extraction and purification. Can 13. Shannon P, Markiel A, Ozier O et al (2003)
J Biochem Physiol 37:911–917 Cytoscape: a software environment for inte-
4. Geier FM, Want EJ, Leroi AM et al (2011) Cross- grated models of biomolecular interaction net-
platform comparison of Caenorhabditis elegans works. Genome Res 13:2498–2504. https://
tissue extraction strategies for comprehensive doi.org/10.1101/gr.1239303
metabolome coverage. Anal Chem 83:3730– 14. Kanehisa M, Goto S (2000) KEGG: Kyoto
3736. https://doi.org/10.1021/ac2001109 encyclopedia of genes and genomes. Nucleic
5. Masson P, Spagou K, Nicholson JK et al (2011) Acids Res 28:27–30
Technical and biological variation in UPLC- 15. Beckonert O, Keun HC, Ebbels TM et al
MS- based untargeted metabolic profiling of (2007) Metabolic profiling, metabolomic and
liver extracts: application in an experimental metabonomic procedures for NMR spectros-
toxicity study on galactosamine. Anal Chem copy of urine, plasma, serum and tissue extracts.
83:1116–1123. https://doi.org/10.1021/ Nat Protoc 2:2692–2703. https://doi.
ac103011b org/10.1038/nprot.2007.376.
6. Anwar MA, Vorkas P, Li JV et al (2015) nprot.2007.376 [pii]
Optimization of metabolite extraction of 16. Nicholson JK, Foxall PJ, Spraul M et al (1995)
human vein tissue for ultra performance liquid 750 MHz 1H and 1H-13C NMR spectros-
chromatography-mass spectrometry and copy of human blood plasma. Anal Chem
nuclear magnetic resonance-based untargeted 67:793–811
metabolic profiling. Analyst 140:7586–7597 17. Yap IK, Brown IJ, Chan Q et al (2010)
7. Vorkas PA, Isaac G, Anwar MA et al (2015) Metabolome-wide association study identifies
Untargeted UPLC-MS profiling pipeline to multiple biomarkers that discriminate north
expand tissue metabolome coverage: applica- and south Chinese populations at differing
tion to cardiovascular disease. Anal Chem risks of cardiovascular disease: INTERMAP
87:4184–4193. https://doi.org/10.1021/ study. J Proteome Res 9:6647–6654. https://
ac503775m doi.org/10.1021/pr100798r
8. Vorkas PA, Shalhoub J, Isaac G et al (2015) 18. Saric J, Wang Y, Li J et al (2008) Species varia-
Metabolic phenotyping of atherosclerotic tion in the fecal metabolome gives insight into
plaques reveals latent associations between free differential gastrointestinal function.
cholesterol and ceramide metabolism in ath- J Proteome Res 7:352–360. https://doi.
erogenesis. J Proteome Res 14:1389–1399. org/10.1021/pr070340k
https://doi.org/10.1021/pr5009898 19. Lofstedt T, Trygg J (2011) OnPLS-a novel
9. Ashrafian H, Li JV, Spagou K et al (2014) multiblock method for the modelling of pre-
Bariatric surgery modulates circulating and car- dictive and orthogonal variation. J Chemom
diac metabolites. J Proteome Res 13:570–580. 25:441–455. https://doi.org/10.1002/
https://doi.org/10.1021/pr400748f cem.1388
10. Anwar MA, Vorkas PA, Li J et al (2016) 20. Veselkov KA, Vingara LK, Masson P et al
Prolonged mechanical circumferential stretch (2011) Optimized preprocessing of ultra-
induces metabolic changes in rat inferior vena performance liquid chromatography/mass
cava. Eur J Vasc Endovasc 52:544–552. spectrometry urinary metabolic profiles for
https://doi.org/10.1016/j.ejvs.2016.07.002 improved information recovery. Anal Chem
11. Vorkas PA, Shalhoub J, Lewis MR et al (2016) 83:5864–5872. https://doi.org/10.1021/
Metabolic phenotypes of carotid atheroscle- ac201065j
Chapter 18
Abstract
The extraction and untargeted UHPLC-HRMS analysis of endogenous steroids in serum samples is
described in this protocol. The employed full-scan acquisition mode provides the adequate sensitivity to
highlight the main endogenous steroids present in blood, including mineralocorticoids, progestogens, and
androgens. Technical aspects for both chromatography and mass spectrometry are discussed in detail,
together with a proposition of setup for sample sequence and data analysis. Furthermore, general com-
ments are given to help the assessment of data quality and system performance.
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_18, © Springer Science+Business Media, LLC, part of Springer Nature 2018
261
262 Federico Ponzetto et al.
2 Materials
Ultrapure water (18.2 MΩ, total organic carbon <5 ppb), organic
solvents (acetonitrile, ACN, and methanol, MeOH), and formic
acid (FA) of HPLC grade or higher purity should be used. The
same quality criteria must be applied to analytical standards to
obtain the highest available purity. Reference endogenous steroid
standards as well as deuterium-labeled internal standards could be
purchased on the market from various providers, such as Sigma-
Aldrich, Steraloids, and Cerilliant. Charcoal stripped human serum
(steroid depleted/negative serum) is purchased from Dunn
Labortechnik GmbH (Asbach, Germany).
2.1 Analytes Stock Endogenous steroids and labeled internal standards should be pur-
Solutions, Internal chased as calibrated stock solutions (1 mg/mL or 100 μg/mL in
Standard, and Quality MeOH or ACN) or as powder. For the latter, stock solutions are
Control Mixtures prepared by weighing the appropriate amount of powder and dis-
solving it in MeOH to obtain a final concentration of 1 mg/
mL. Stock solutions are finally diluted in MeOH to obtain
intermediate solutions at a concentration of 10 μg/mL. Both stock
Steroidomics 263
3 Methods
3.1 Serum Sample Supported liquid extraction (SLE) on ISOLUTE® SLE+ 400 μL
Extraction 96-well plates (Biotage, Uppsala, Sweden) is used to extract ste-
roid hormones from serum samples. The extraction procedure
consists of five different steps:
1. For each sample, an aliquot of 200 μL of serum is spiked with
20 μL of the IS-mix, diluted with 200 μL of water, and then
agitated for 15 min at 250 rpm.
2. Each well is then loaded with 400 μL of the previously diluted
sample, and a positive pressure of 3 psi is applied for 30 s to
facilitate sample loading and adsorption.
3. The elution is carried out, after a 5 min waiting period, by add-
ing 700 μL of dichloromethane to each well and applying a
pressure of 3 psi for 1 min.
4. Extracts are then collected in collection plates equipped with
1.5 mL glass inserts, evaporated to dryness for approximately
15 min at 40 °C under a gentle stream of nitrogen, and finally
reconstituted with 100 μL of a MeOH-H2O 50:50 (v/v) mix-
ture (reconstitution solvent).
5. After 15 min of gentle shaking (250 rpm), 10 μL of each extract
is injected into UHPLC-HRMS system for analyses.
3.4 HRMS Prior to the injection of the extracted serum samples, it is recom-
Maintenance mended to carry out the cleaning of the ESI source followed by
and Calibration the calibration of the MS system. The MS cleaning procedure
involves the cone and the ion transfer tube that are removed from
the MS instrument after cooling the ESI source temperature to
room temperature. The instrument is then switched on standby
mode, and the two parts of the ion source are subjected to three
washing steps of 10 min sonication each in three different
solvents:
1. 10% FA in ultrapure H2O.
2. Ultrapure H2O.
3. HPLC grade MeOH.
Once the third step is completed, the cone and the ion transfer
tube are dried under a steam of argon and then assembled again on
the instrument (see Note 7). Then, the calibration MS tune file
(sheath gas flow rate set to 3 arbitrary units, both auxiliary and
sweep gas flow set to 0; ion spray voltage of 3.5 kV and capillary
temperature of 320 °C with the S-Lens RF level at 50%) is loaded
and the ESI source connected to the syringe filled up with the
Positive Ion Calibration standard mixture. The flow rate is set at
5 μL/min, and the system should be stabilized for 10 min; the cali-
bration is performed only when the TIC variation is constantly
lower than 10%.
General
Runtime 1 to 17 min
Polarity positive
In-source CID 0.0 eV
Default charge state 1
Inclusion -
Exclusion -
Tags -
Full MS
Microscans 1
Resolution 70,000
AGC target 3e6
Maximum IT 125 ms
Number of scan range 1
Scan range 200 to 600 m/z
Spectrum data type Profile
dd-MS2 / dd-SIM
Microscans 1
Resolution 17,500
AGC target 5e4
Maximum IT 64 ms
Loop count 5
MSX count 1
TopN 5
Isolation window 0.4 m/z
Isolation offset 0.0 m/z
Fixed first mass -
NCE / stepped NCE 20, 40, 60
Spectrum data type Profile
dd Settings
Minimum AGC target 5.00e3
Intensity threshold 7.8e4
Apex trigger 2 to 6 s
Charge exclusion -
Peptide match -
Exclude isotopes -
Dynamic exclusion 6.0 s
RT: 9.39 - 9.80 SM: 7G RT: 10.87 - 11.21 SM: 7G RT: 11.16 - 11.55 SM: 7G RT: 9.95 - 10.18 SM: 7G
RT: 9.56 NL: 3.02E6 RT: 11.05 NL: 7.76E5 RT: 11.36 NL: 7.00E5 RT: 10.07 NL: 5.12E5
MA: 7228687 m/z= MA: 1863896 m/z= MA: 1725832 m/z= MA: 1100718 m/z=
100 289.21332-289.21910 100 273.21856-273.22402 100 273.21856-273.22402 100 287.19769-287.20343
F: FTMS + p ESI Full F: FTMS + p ESI Full F: FTMS + p ESI Full F: FTMS + p ESI Full
ms [200.00-600.00] ms [200.00-600.00] ms [200.00-600.00] ms [200.00-600.00]
MS MS MS MS
Relative Abundance
Relative Abundance
Relative Abundance
Relative Abundance
50 50 50 50
9.97
0 0 0 0
9.4 9.5 9.6 9.7 9.8 10.9 11.0 11.1 11.2 11.2 11.3 11.4 11.5 10.0 10.1
Testosterone Etiocholanolone Androsterone Androstenedione
RT: 10.59 - 10.90 SM: 7G RT: 9.99 - 10.28 SM: 7G RT: 6.92 - 7.20 SM: 7G RT: 7.94 - 8.29 SM: 7G
RT: 10.74 NL: 1.08E5 RT: 10.13 NL: 9.61E5 RT: 7.05 NL: 1.11E7 RT: 8.11 NL: 2.93E6
MA: 255613 m/z= MA: 2195338 m/z= MA: 22752916 m/z= MA: 6375086 m/z=
100 291.22895-291.23477 100 271.20293-271.20835 100 361.19734-361.20456 100 347.21822-347.22516
F: FTMS + p ESI Full F: FTMS + p ESI Full F: FTMS + p ESI Full F: FTMS + p ESI Full
ms [200.00-600.00] ms [200.00-600.00] ms [200.00-600.00] ms [200.00-600.00]
MS MS MS MS
Relative Abundance
Relative Abundance
Relative Abundance
Relative Abundance
50 50 50 50
0 0 0 0
10.6 10.7 10.8 10.9 10.0 10.1 10.2 7.0 7.1 8.0 8.1 8.2
DHT DHEA Cortisone Corticosterone
RT: 6.75 - 7.11 SM: 7G RT: 8.20 - 8.45 SM: 7G RT: 10.02 - 10.44 SM: 7G RT: 12.16 - 12.39 SM: 7G
RT: 6.92 NL: 6.04E7 RT: 8.32 NL: 1.53E5 RT: 10.19 NL: 5.52E5 RT: 12.27 NL: 1.56E5
MA: 125368564 m/z= MA: 339825 m/z= MA: 1300417 m/z= MA: 422340 m/z=
100 363.21297-363.22023 100 347.21822-347.22516 100 331.22346-331.23008 100 315.22871-315.23501
F: FTMS + p ESI Full F: FTMS + p ESI Full F: FTMS + p ESI Full F: FTMS + p ESI Full
ms [200.00-600.00] ms [200.00-600.00] ms [200.00-600.00] ms [200.00-600.00]
MS MS MS MS
Relative Abundance
Relative Abundance
Relative Abundance
Relative Abundance
50 50 50 50
0 0 0 0
6.8 6.9 7.0 7.1 8.2 8.3 8.4 10.1 10.2 10.3 10.4 12.2 12.3
Cortisol 11-deoxycortisol 17α-hydroxyprogesterone Progesterone
Chemical formula, retention time, and most abundant ion m/z value of the 101 endogenous steroids of the in-house steroid database
(continued)
269
Table 1
270
(continued)
dione
3α,5α-Tetrahydrocorticosterone C21H34O4 350.24571 8.46 315.23186
3α,5α-Tetrahydrocortisol C21H34O5 366.24063 7.41 331.22677
3α,5β-Tetrahydroaldosterone C21H32O5 364.22498 6.54 387.21420
3α,5β-Tetrahydrocorticosterone C21H34O4 350.24571 8.34 315.23186
3α,5β-Tetrahydrocortisol C21H34O5 366.24063 7.45 331.22677
3α,5β-Tetrahydrocortisone C21H32O5 364.22498 7.77 347.22169
5α,20α-Tetrahydroprogesterone C21H34O2 318.25588 12.47 301.25259
5α-Androstane-3α,17β-diol C19H32O2 292.24023 10.61 257.22638
5α-Androstan-3β-ol-7,17-dione C19H28O3 304.20385 7.36 287.20056
5α-Androstane-3β,12β,15α-triol C19H32O3 308.23515 5.69 273.22129
5α-Androstane-3β,7α,16β-triol C19H32O3 308.23515 6.73 273.22129
5α-Androstanedione C19H28O2 288.20893 11.35 289.21621
5α-Androstane-3β,17β-diol C19H32O2 292.24023 9.97 275.23694
Most abundant ion [m/z]
Chemical Retention time
Compound name formula Exact mass [Da] [min] [M+H]+ [M+Na]+ [M–H2O]+ [M–2H2O]+
5α-Dihydro-11-deoxycorticosterone C21H32O3 332.23515 10.98 333.24242
5α-Dihydrocorticosterone C21H32O4 348.23006 8.98 349.23734
5α-Dihydrocortisol C21H32O5 364.22498 7.54 365.23225
5α-Dihydrocortisone C21H30O5 362.20933 7.58 363.21660
5α-Dihydroprogesterone C21H32O2 316.24023 13.47 317.24751
5-Androsten-3β,17β-diol-16-one C19H28O3 304.20385 7.31 287.20056
5β-Androstane-3α,17β-diol C19H32O2 292.24023 10.36 275.23694
5β-Androstan-11α-ol-3,17-dione C19H28O3 304.20385 8.53 269.18999
5β-Androstan-11β-ol-3,17-dione C19H28O3 304.20385 9.13 305.21112
5β-Androstane-3α,17α-diol C19H32O2 292.24023 11.57 257.22638
5β-Androstane-3β,17α-diol C19H32O2 292.24023 10.74 257.22638
5β-Androstanedione C19H28O2 288.20893 11.45 289.21621
5β-Androstane-3β,17β-diol C19H32O2 292.24023 10.08 275.23694
5β-Dihydro-11-dehydrocorticosterone C21H30O4 346.21441 8.87 347.22169
5β-Dihydrocorticosterone C21H32O4 348.23006 9.02 349.23734
5β-Dihydrocortisol C21H32O5 364.22498 7.86 347.22169
5β-Dihydrocortisone C21H30O5 362.20933 8.10 363.21660
Steroidomics
(continued)
271
Table 1
272
(continued)
3.7 Analytical
The analytical sequence should be constructed carefully with the
Sequence
aim of simultaneously providing the best compromise between a
satisfying data acquisition and a clear measure of the data quality
and system performance. Solvent blank, negative serum samples,
conditioning samples, and QC-mix samples are added to the ste-
roidomic study serum sample injections helping the assessment of
the analysis quality. The proposal of a steroidomic analytical
sequence is described below:
1. A solvent blank injection at the beginning of the analytical
sequence helps the identification of background noise related to
the reconstitution solvent. If more than one solvent blank injec-
tion is performed, it is also possible to evaluate the presence of
any carry over effect.
2. Before starting the conditioning process, a minimum of two
injections of negative serum samples should be performed.
These injections are useful in evaluating the status of the ana-
lytical system, in particular in regard to the chromatographic
column. Indeed, the injection of negative serum samples could
help the assessment of any carry over of endogenous steroids
related to previous analyses, which is difficult to detect when
injecting the solvent blank.
3. After the negative serum sample injections, a minimum of five
QC-mix sample injections should be performed to stabilize the
chromatographic system. Furthermore, it allows conditioning
the HRMS system, because of a significant performance decrease
across the first injections of serum matrix. When QC-mix vol-
ume is limited due to scarce volume of study samples, it is also
possible to perform the system conditioning by injecting other
serum samples; the key aspect is to inject matrix-based samples
that are as similar as possible to the serum samples of the study.
For example, if the study is conducted on male subjects, it could
be possible to use, as conditioning samples, any other male
serum samples or a commercially available pool of male serum.
The system conditioning is mandatory to obtain reliable results
with adequate repeatability.
4. QC-mix sample injections are of crucial importance for the sta-
tistical analysis and for the evaluation of the instrument perfor-
mance in terms of detection and sensitivity of the analytes of
interest. The analysis of steroidomic study samples should
always start and end with a QC-mix sample injection to enable
quality assessment at the beginning and end of the analytical
batch; QC-mix injections could also be used during the data
treatment for normalization purposes. As a minimum of five
QC-mix injections are mandatory to obtain a sufficient robust-
ness for the statistical analysis, these injections should be per-
Steroidomics 275
3.8 Data Treatment Raw data obtained from the UHPLC-HRMS analysis are pro-
cessed with appropriate software to obtain peak areas of all peaks
detected in the steroidomic study samples. Several solutions and
commercial and open-source-based strategies could be engaged;
however, the following steps should be achieved:
1. The first step of the data treatment is the alignment of the chro-
matograms. In most of the cases, it is possible to choose between
an automatic or manual alignment, in which the user should
define the peak of interest that should be used to align the chro-
matograms. We suggest, in the case of untargeted analyses,
where it is not possible to assess the nature of all the peaks in the
chromatogram and then decide which are important and which
are not, to use the automatic alignment, which uses various
peaks as a reference for the alignment.
2. The second and probably one of the most important steps of
the data treatment concerns the so-called peak picking. Here,
the user has to define where, and how, the software should
search for peaks in the aligned chromatograms. It could be
appropriate to perform the peak picking only in the retention
time window covering the period of the gradient, i.e., to exclude
all compounds that are too well retained and elute only in the
washing step or those which are not retained at all hence and
elute during the dead time. It is also necessary to define a sensi-
tivity threshold (the higher the sensitivity, the larger the num-
ber of peaks detected) and the type of adducts. For this step, we
suggest using the highest sensitivity in order to detect the maxi-
mum number of peaks, which could then be reduced employ-
ing statistical tools, and to include only the adducts typical of
276 Federico Ponzetto et al.
4 Notes
References
1. Sanderson JT (2006) The steroid hormone reference intervals in healthy adults. Steroids
biosynthesis pathway as a target for endocrine- 76:244–253
disrupting chemicals. Toxicol Sci 94:3–21 9. Rosner W, Auchus RJ, Azziz R et al (2007)
2. Arukwe A (2008) Steroidogenic acute regula- Position statement: utility, limitations, and pit-
tory (StAR) protein and cholesterol side- falls in measuring testosterone: an endocrine
chain cleavage (P450scc)-regulated society position statement. J Clin Endocrinol
steroidogenesis as an organ-specific molecu- Metab 92:405–413
lar and cellular target for endocrine disrupt- 10. Handelsman DJ, Wartofsky L (2013)
ing chemicals in fish. Cell Biol Toxicol Requirement for mass spectrometry sex steroid
24:527–540 assays in the journal of clinical endocrinology
3. Shackleton CHL (2012) Role of a disordered and metabolism. J Clin Endocrinol Metab
steroid metabolome in the elucidation of sterol 98:3971–3973
and steroid biosynthesis. Lipids 47:1–12 11. Wartofsky L, Handelsman DJ (2010)
4. Kicman AT (2008) Pharmacology of anabolic Standardization of hormonal assays for the
steroids. Br J Pharmacol 154:502–521 21st century. J Clin Endocrinol Metab
5. World Anti-Doping Agency (WADA), 95:5141–5143
Montreal (2015) Anti-doping testing figures 12. World Anti-Doping Agency (WADA),
report. http://www.wada-ama.org. Accessed Montreal (2016) endogenous anabolic andro-
May 2017 genic steroids, measurement and reporting,
6. Basaria S (2010) Androgen abuse in athletes: technical document TD2016EAAS. http://
detection and consequences. J Clin Endocrinol www.wada-ama.org. Accessed May 2017
Metab 95:1533–1543 13. Sjovall J (2004) Fifty years with bile acids and
7. Büttner A, Thieme D (2010) Side effects of steroids in health and disease. Lipids
anabolic androgenic steroids: pathological 39:703–722
findings and structure-activity relationships. 14. Randazzo GM, Tonoli D, Strajhar P et al
Handb Exp Pharmacol 195:459–484 (2017) Enhanced metabolite annotation via
8. Fanelli F, Belluomo I, Di Lallo VD et al (2011) dynamic retention time prediction: steroido-
Serum steroid profiling by isotopic dilution– genesis alterations as a case study. J Chromatogr
liquid chromatography–mass spectrometry: B Analyt Technol Biomed Life Sci.
comparison with current immunoassays and 1071:11–18
Chapter 19
Abstract
Metabolomics can be of great value in the study of exercise metabolism. However, because of the high
intraindividual and interindividual biological variability of the human metabolome, special considerations
should be taken into account when designing an acute-exercise metabolomic study. To study different
exercise parameters, e.g., different exercise modes, intensities, etc., a crossover study design, where each
participant acts as their own control, is preferable to a parallel design, one involving different groups of
participants. Moreover, the study should include a no exercise, control trial. Before each trial, participants
should follow carefully designed preparatory steps to control for possible confounding factors, i.e., main-
tain repeatable and constant conditions for all individual trials of the study to minimize variation due to
factors other than the one(s) being studied. This chapter focuses on the design of human metabolomic
studies, where the intervention is an acute metabolic challenge, such as an exercise bout or a test meal, and
presents some basic steps for screening potential participants, performing preliminary tests, preparing for
the trial day, and performing the trial.
1 Introduction
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0_19, © Springer Science+Business Media, LLC, part of Springer Nature 2018
279
280 Aikaterina Siopi and Vassilis Mougios
2 Materials
3 Methods
3.2 Preliminary Tests After the screening process and before the trials, participants usu-
ally need to go through some preliminary tests (see Note 4). These
can include:
1. Body composition measurements, such as lean body mass, total
fat mass, and visceral fat mass (see Note 5). Make sure the par-
ticipants have followed the necessary preparations for the
respective analysis. These measurements can be used as descrip-
tive characteristics of the study sample. Moreover, they can be
correlated with outcomes of the metabolomic analysis.
2. Measurement or estimation of resting energy expenditure (see
Note 6). This measurement will be used to design the dietary
plan of the participants during the study in order to decrease
intraindividual and interindividual variations.
3. Cardiorespiratory fitness assessment. Perform a maximal or
submaximal incremental test (depending on your study sample)
to assess maximal oxygen consumption (VO2max) and maximal
heart rate [12]. This measurement may be used to set the exer-
cise intensity for endurance exercise trials.
4. Muscular strength measurement. Perform a determination or
prediction (depending on your study sample) of one-repetition
maximum [13]. This measurement may be used to set the exer-
cise intensity for resistance exercise trials.
3.3 Preparation Arrange some extra time at the end of the preliminary tests appoint-
for the Exercise Trials ment to talk with each participant about the scheduling and prepa-
ration for the trials. Again, be thorough and provide all instructions
in writing as well as verbally for the participants to take home (see
Note 2). Schedule the next appointment (see Notes 4 and 7). A
Design of Acute-Exercise Metabolomic Studies 283
Fig. 1 Overview of the preparation for each trial of the study (see Note 10)
3.4 Trial Day After arrival of the participant to the lab and before starting the
trial:
1. Weigh them to make sure that there has been no considerable
change since the previous measurement.
2. Go with them through a checklist to make sure they have suc-
cessfully followed all necessary steps of the preparation
process.
3. Check their dietary and physical activity records of the preced-
ing days.
4. Take a 24-h dietary recall to cross-validate the dietary record of
the previous day and to make sure it has been completed prop-
erly (see Note 11).
5. If they have failed to follow any of the preparation steps and you
feel that the reliability of the process has been compromised,
you may have to reschedule the trial.
During the trial:
1. Have the participants remain seated and relaxed during the
non-exercising parts of the trial.
2. Mark their water consumption during the first trial and have
them replicate it during the next trial(s).
Design of Acute-Exercise Metabolomic Studies 285
4 Notes
hot tub for the last 24 h; and abstention from diuretics for the
last 7 days prior to the analysis.
6. If possible, use the method of indirect calorimetry with the
canopy technique to measure resting energy expenditure.
Alternatively, you can use equations to estimate energy needs
[16].
7. In a crossover design, each participant must complete all trials
in a randomized order. You can use a random-number genera-
tor software (one of the many available online for free) to
obtain random sequences.
8. In human trials, the interpersonal communication skills of the
researcher/staff are very important.
9. The dietary plan should be carefully designed to maximize
compliance and avoid major effects of diet on the metabo-
lome. It should not include complicated recipes, expensive
ingredients, or food that the participant does not like, is not
used to, or is allergic to. Ideally, the research project should
provide catering for all meals of the participants.
10. Depending on how many sampling points there are through-
out the day of each trial, you may need to control diet and
physical activity on the day of the trial as well.
11. Dietary recalls need to be taken by a trained and experienced
professional to be accurate.
12. The best way to match acute-exercise bouts is to measure
exercise energy expenditure by indirect calorimetry.
References
A D
Acute-exercise������������������������������������������������������������������279 Data analysis workflow��������������������������������������� 27, 103, 232
Algorithm���������������30–32, 34, 36, 37, 42, 112, 142–144, 251 Data conversion�������������������������������������������29, 35, 36, 43, 45
Analyte quantitation�������������������� 21, 27, 66, 78, 84, 209, 225 Data integration����������������������������������������� 35, 240, 245, 256
Analytical batch���������������������������������������20, 94, 95, 274, 277 Data modeling��������������������������������������������������������������������28
Androgens���������������������������������������������������������������� 261, 267 Data quality assessment�������������������������������������� 96, 100, 103
Anionic metabolite����������������������������184, 185, 187, 188, 190 Data treatment��������������������������������������� v, 10, 27–38, 78–79,
Annotation���������������������� 41, 43, 46, 220, 232, 254, 258, 276 246–248, 251–255, 263, 274–276
Aroma�������������������������������������������������������� 48, 178, 214–216 Data visualization���������������������������������������������������������������96
Artificial chewing�����������������������������������������������������216–219 Deconvolution������������� 84, 232, 233, 243, 245, 251, 252, 254
Atmospheric-pressure chemical ionization Derivatization������������������������� 7, 84, 135, 139, 140, 142–144,
(APCI)�������������������������������������������������������������9, 214 146, 151, 156, 159, 164–173, 262
Authentication������������������������������������������������������������������209 Design of experiment (DoE)���������������������������� v, 27, 35, 134,
Authenticity����������������������������������������������������������������� v, 228 216, 227, 228, 231, 233
Direct infusion mass spectrometry (DIMS)�����������������7, 214
B Dispersive liquid liquid microextraction
Background electrolyte (BGE)���������������������������������185–189 (DLLME)���������������������������� 159–162, 164–173, 176
Biological database�������������������������������������������������������������42
E
Biological interpretation����������������������������� 41, 245, 255, 256
Biological variability������������������������������������������������� 216, 226 Electrospray ionization (ESI)��������������� 9, 10, 18, 91, 95, 102,
Blood��������������������������������� v, 7, 15, 66, 70, 77, 118, 119, 121, 112, 184, 186, 190, 227, 229–231, 233, 242, 264, 265
122, 125, 126, 130, 136, 138–139, 146, 280, 282 Endocrine diseases������������������������������������������������������������261
Endogenous metabolites��������������������������������� 7, 94, 183, 184
C Equilibration injections������������������������������������������ 77, 78, 80
Caenorhabditis elegans��������������������������������������������������������240 Exact mass�������������������������������������������������� 42–44, 46–49, 53
Calibration curve����������������������������������������������������������77–80 Exercise metabolomics���������������������������������������������279–281
Calibration solutions���������������������������17, 101, 105–106, 175 Exercise modes�����������������������������������������������������������������280
Capillary electrophoresis (CE)������������ v, 9, 83, 188, 189, 250 External calibration curve���������������������������������������������77–79
Carr-Purcell-Meiboom-Gill sequence (CPMG)������������126, Extracted ion chromatogram (XIC)�����������������������������22–24
127, 249, 254 Extraction������������������� 5, 27, 35, 70, 79, 92, 93, 100–102, 104,
Cationic metabolite������������������������������������������ 187, 188, 190 105, 114, 119, 121–124, 129, 134, 136–139, 145, 146,
Cell culture���������������������������������������125, 129, 133, 136–138, 150, 151, 153, 154, 156, 161, 176, 196, 199, 201, 204,
140, 141, 162, 190 207, 208, 220, 222, 230, 233, 240, 241, 245–247, 251,
Cerebrospinal fluid (CSF)�������������������������������� 118, 125, 130 254, 256–258, 262–264, 267, 276
Chemical structure������������������������������������������� 42, 46, 47, 54
F
Chemoinformatics�����������������������������������������������������v, 41–60
Chloroformate derivatization�������������������������������������������160 Features���������������������21, 23, 24, 28, 30, 33–36, 38, 112, 113,
Chromatogram����������������������� 8, 21–24, 93, 95, 96, 107, 108, 135, 140, 160, 226, 240, 252, 253, 255, 265, 276, 277
140, 141, 151, 197, 227, 248, 249, 267, 275 Fecal sample�������������������������������������������������������������149–156
Chromatogram alignment������������������������������������������ 21, 275 Food����������������������������������������������v, 7, 11, 70, 203, 205, 213,
Command line��������������������������������� 30, 43, 44, 46, 48, 53, 60 214, 216, 284, 286
Confounding factors����������������������������������� 29, 280, 281, 283 Food analysis������������������������������������������������������������ 199, 203
Correlation analysis�������������������������������������������������� 144, 256 Fresh fruit tissue����������������������������������������������� 215, 217, 218
Georgios A. Theodoridis et al. (eds.), Metabolic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1738,
https://doi.org/10.1007/978-1-4939-7643-0, © Springer Science+Business Media, LLC, part of Springer Nature 2018
289
Metabolic Profiling: Methods and Protocols
290 Index
G Metabolic profiling���������� v, 3–12, 84, 99–101, 106, 112, 113,
117–130, 134, 150, 151, 185–188, 190, 240, 285
Gas chromatography (GC)����������������������v, 3, 4, 7, 10, 15, 16, Metabolome coverage���������������������������������������� 99, 239, 245,
83, 99, 134, 150–152, 154–156, 159–162, 164–173, 249, 250, 253
176, 196, 217, 220, 226, 261 Metabolomics�������������������������� v, 3, 15, 23, 24, 35, 41, 44, 54,
Global metabolic profiling�����������������������������������������v, 16, 17 58, 65, 117, 133, 149–156, 159–179, 183, 195, 203,
Gut microbiota�����������������������������������������������������������������150 205, 225, 239–259, 279–286
Metabonomics�����������������������������v, 3, 5, 15, 83, 84, 239, 245,
H
249, 250, 253
Headspace������������������������������������������������ 215, 216, 218–222 Metabotype���������������������������������������������������������������� 4, 5, 15
Heptafluorobutyl chloroformate (HFBCF)������������ 159–162, Metadata organization������������������������������������������ 29–30, 234
164–173, 176 Method validation���������������������������������������������� 16, 225, 226
Heteronuclear single quantum correlation Methoximation�����������������������������������������������������������������135
(HSQC)���������������������������������������������������������������128 Mineralocorticoids�����������������������������������������������������������267
High-resolution magic-angle spinning (HR-MAS)�����������204 Missing peaks����������������������������������������������� 29, 34, 142, 232
Human metabolome������������������������������11, 55, 145, 255, 280 Molecular formula���������������������������������10, 42–45, 49–51, 53
Human metabolome database (HMDB)��������������� 11, 42, 55, Mono-layer extractions�����������������������������������������������������240
79, 113, 145, 209, 255, 276 Multi-method�������������������������������������������������������������������240
Hydrophilic interaction liquid chromatography
(HILIC)�������������������������8, 66, 84, 100, 101, 104, 105, N
107, 109–111, 114, 152, 154–156, 227, 229–231, 233, Normalization�������������������������� 16, 20, 21, 41, 124, 135–138,
240, 242, 245, 247, 250, 252 140–144, 175, 179, 232, 252, 274
Nuclear magnetic resonance (NMR)����������������v, 4–7, 9–11, 15,
I
65, 83, 99, 117, 134, 150–155, 195, 197, 200, 203–210,
Identification������������������������� v, 4, 5, 7, 10, 21, 30, 41, 42, 49, 240, 242, 243, 245, 246, 249, 254, 255, 257, 258
53, 66, 92, 93, 99, 102, 109, 113, 128, 134, 136, 137, Nuclear Overhauser effect spectroscopy
140, 141, 196, 203, 209, 252, 255, 265, 274, 276, 280 (NOESY )����������������������������������������������������� 127, 130
In-house database������������������������������������� 265, 267–273, 276 Nutrimetabolomics�����������������������������������������������������������118
Ion pair chromatography���������������������������������������� 66, 83–96
Isotope pattern������������������������������������������������������� 42, 49–53 O
Oenological topic�����������������������������������������������������227–228
J
Online databases���������������������������������������������������������������276
Java������������������������������������������������������������������� 43–46, 48, 60 Orthogonal projection to latent structures
(OPLS)����������������������������������������� 210, 243, 253, 255
K
P
Kyoto Encyclopedia of Genes and Genomes
(KEGG)���������������� 11, 42, 56–60, 145, 243, 255, 256 Participant����������������������������������������������������������������280–286
Pathway mapping��������������������������������������������� 243, 255, 256
L Peak alignment�������������������������������������������9, 21, 23, 41, 102,
Large-scale monitoring����������������������������������������������������261 112, 232, 275
LipidMaps�����������������������������������������������42, 55, 60, 113, 276 Peak picking����������������������������� 18, 22, 29–31, 34, 36, 37, 41,
Liquid chromatography (LC)�������������������� 18, 27–38, 66–69, 112, 232, 233, 251, 275, 276
85, 100, 107 Pharmacometabolomics����������������������������������������������������118
LogD����������������������������������������������������������������������������46–49 Phase I metabolites�����������������������������������������������������������267
Physicochemical parameter������������������������������������������������43
M Plants������������������������������������������������������������ 7, 136, 146, 234
Pre-analytical considerations�������������������������������������� 92, 134
Mass spectrometry (MS)�������������������� v, 4, 7–10, 15, 16, 23,
Preliminary tests������������������������������������������������������� 140, 282
24, 27–38, 46, 49, 54, 65–80, 83, 84, 91–92, 99–101,
Principal component analysis (PCA)����������������� 9, 10, 21–24,
104, 107–109, 112, 113, 115, 118, 134, 137, 140,
96, 144, 209–210, 228, 232, 233, 235, 243, 252, 253,
141, 150, 183–187, 190, 191, 195, 203, 213, 218,
255, 277
220, 226, 229–231, 240, 242, 243, 245, 248, 251,
Progestogens���������������������������������������������������������������������267
255, 262, 264–266, 276
Proton transfer reaction-mass spectrometry
Matrix effect������������������������������������������������� 66, 79, 135, 226
(PTR-MS)������������������������������������������������v, 214, 215
Metabolic Profiling: Methods and Protocols
Index
291