You are on page 1of 9

Journal of Chromatography B, 1053 (2017) 72–80

Contents lists available at ScienceDirect

Journal of Chromatography B
journal homepage: www.elsevier.com/locate/chromb

Lipidomics by ultrahigh performance liquid chromatography-high


resolution mass spectrometry and its application to complex
biological samples
Alexander Triebl a , Martin Trötzmüller a,∗ , Jürgen Hartler b , Tatjana Stojakovic c ,
Harald C. Köfeler a,d
a
Core Facility for Mass Spectrometry, Center for Medical Research, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria
b
Institute of Computational Biotechnology, Graz University of Technology, Petersgasse 14, 8010 Graz, Austria
c
Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
d
Omics Center Graz, Stiftingtalstrasse 24, 8010 Graz, Austria

a r t i c l e i n f o a b s t r a c t

Article history: An improved approach for selective and sensitive identification and quantitation of lipid molecular
Received 29 November 2016 species using reversed phase chromatography coupled to high resolution mass spectrometry was devel-
Received in revised form 8 March 2017 oped. The method is applicable to a wide variety of biological matrices using a simple liquid-liquid
Accepted 22 March 2017
extraction procedure.
Available online 24 March 2017
Together, this approach combines multiple selectivity criteria: Reversed phase chromatography sep-
arates lipids according to their acyl chain length and degree of unsaturation and is capable of resolving
Keywords:
positional isomers of lysophospholipids, as well as structural isomers of diacyl phospholipids and glyc-
High-resolution mass spectrometry
High performance liquid chromatography
erolipids. Orbitrap mass spectrometry delivers the elemental composition of both positive and negative
Quantitative lipidomics ions with high mass accuracy. Finally, automatically generated tandem mass spectra provide structural
Lipid structural isomers insight into numerous glycerolipids, phospholipids, and sphingolipids within a single run.
Calibration showed linearity ranges of more than four orders of magnitude, good values for accuracy
and precision at biologically relevant concentration levels, and limits of quantitation of a few femtomoles
on column.
Hundreds of lipid molecular species were detected and quantified in three different biological matrices,
which cover well the wide variety and complexity of various model organisms in lipidomic research.
Together with a software package, this method is a prime choice for global lipidomic analysis of even the
most complex biological samples.
© 2017 Elsevier B.V. All rights reserved.

1. Introduction

The general term “lipids” is used to describe a plethora of


biomolecules that differ in structure as well as in function, rang-
ing from simple fatty acids to complex saccharolipids and from
use in energy storage to signal transduction and as vitamins [1,2].
Abbreviations: Cer, ceramide; CL, cardiolipin; DG, diacylglycerol; EIC, Lipidomics is the general study of lipids and their contributions to
extracted ion chromatogram; FTICR, fourier transform ion cyclotron reso- both physiological processes and pathological conditions, such as
nance; FWHM, full width half maximum; HexCer, hexosyl ceramide; HPLC, cancer and diabetes [3].
high performance liquid chromatography; LC/MS, liquid chromatography mass
The two approaches in lipidomic analyses are shotgun tech-
spectrometry; LLOQ, lower limit of quantitation; LPC, lysophosphatidylcholine;
LPE, lysophosphatidylethanolamine; MS/MS, tandem mass spectrometry; MTBE, niques and separation techniques [4]. Shotgun techniques rely on
methyl tert butyl ether; PA, phosphatidic acid; PC, phosphatidylcholine; PE, stable infusion of sample into the ion source without prior sep-
phosphatidylethanolamine; PG, phosphatidylglycerol; PI, phosphatidylinositol; PIP, aration and subsequent detection by either high-resolution mass
phosphatidylinositolphosphate; PS, phosphatidylserine; SFC, supercritical fluid spectrometers [5–12] or a combination of tandem mass spectrom-
chromatography; SM, sphingomyelin; TG, triacylglycerol; UHPLC, ultrahigh perfor-
etry (MS/MS) scans [13–15] to identify lipids by their mass and/or
mance liquid chromatography; ULOQ, upper limit of quantitation.
∗ Corresponding author. characteristic head group and chain fragments. Similar techniques
E-mail address: martin.troetzmueller@medunigraz.at (M. Trötzmüller). are intrasource separation [16], where addition of modifiers causes

http://dx.doi.org/10.1016/j.jchromb.2017.03.027
1570-0232/© 2017 Elsevier B.V. All rights reserved.
A. Triebl et al. / J. Chromatogr. B 1053 (2017) 72–80 73

selective ionization of lipid classes, and ion-mobility mass spec- grade) were purchased from Sigma-Aldrich (St. Louis, MO, USA);
trometry, where the physicochemical properties of ions can be used chloroform and formic acid were purchased from Merck (Darm-
to separate lipid classes and structural or even positional isomers stadt, Germany). Deionized water was obtained from an in-house
[17–23]. MilliQ Gradient A10 system (Millipore, Billerica, MA, USA).
While shotgun methods are an excellent tool for high-
throughput profiling, ion suppression effects may limit their ability 2.2. Lipid standards
to detect low-abundance components, and the lack of separation
prior to mass analysis may diminish the informative value of MS/MS Lipid standards used for quantification and validation (listed
spectra. Adducts of lipids belonging to different classes, or their iso- in Supplementary Tables 2 and 3, respectively) were purchased
topes, may have the same nominal mass which cannot be resolved from Avanti Polar Lipids (Alabaster, AL, USA) and from Larodan Fine
during MS/MS isolation, leading to an MS/MS spectrum with sig- Chemicals AB (Malmö, Sweden).
nals from multiple precursor lipids [24]. Also, some labile lipids,
such as phosphatidylserine [25–27] or cerebrosides (unpublished 2.3. Lipid extraction
observation), may undergo spontaneous in-source fragmentation,
artificially forming ions of phosphatidic acid (PA) or ceramides, The MTBE based lipid extraction protocol employed is a mod-
respectively. Chromatography causes these lipids to elute at dif- ified version of the original extraction protocol published by
ferent retention times so that the artificially generated ions can be Matyash et al. [32]. Methanol (1.5 mL) and MTBE (5 mL) were added
clearly discriminated from the authentic sample components. to the samples (50 mg murine liver, 100 mg of C. elegans embryos,
Furthermore, separation of lipids has the advantage of minimiz- or 106 A549 adenocarcinomic human alveolar basal epithelial cells,
ing the negative influence of interfering compounds in terms of ion respectively) in 12 mL glass tubes with teflon lined caps and the
suppression and mixed MS/MS spectra. Ultrahigh performance liq- mixture was incubated for 10 min in an overhead shaker at room
uid chromatography (UHPLC), the evolution of conventional high temperature. After addition of 1.25 mL deionized water and 10 min
performance liquid chromatography (HPLC) brought by the advent of additional shaking, the mixture was centrifuged for 5 min at 1
of sub-2 m particle columns, is the most widespread separation 350 × g and the upper phase was transferred to a new glass tube.
method. Additionally, core-shell columns are capable of delivering The lower phase was re-extracted with 2 mL of the upper phase
performance comparable to UHPLC at HPLC pressures [28]. Super- of MTBE/methanol/deionized water (10:3:2.5, v/v/v) and again the
critical fluid chromatography (SFC) is a method with promising upper phase was collected, combined with the upper phase from
potential for lipidomic analyses providing shorter analysis times the first extraction, evaporated in a vacuum centrifuge (Thermo
and highest chromatographic resolution [29,30]. Fisher Scientific, Waltham, MA, USA) and dissolved in chloro-
The method previously developed in our lab [31], while at the form/methanol (1:1, v/v) for storage at −20 ◦ C. The liver extract was
time able to generate unprecedented data quality, still suffered dissolved in a volume of 30 mL, the C. elegans extract in 500 ␮L and
from some drawbacks, which collectively made it unsuitable for the A549 cell extract in 1 mL to account for the varying lipid content.
analysis of highly complex lipidomes. Firstly, use of different HPLC Prior to analysis, the internal standards were added (see Section
mobile phases for positive and negative polarity caused lipids to 2.6), the storage solvent was evaporated under a gentle stream
elute at different retention times in positive and negative mode, of nitrogen and the sample reconstituted in the same volume of
complicating data analysis. Secondly, the chromatographic reso- injection solvent isopropanol/chloroform/methanol (90:5:5, v/v/v).
lution and separation power proved to be insufficient for more
sophisticated lipidomic samples. Additionally, the hybrid ion trap- 2.4. LC method
Fourier transform ion cyclotron resonance (FTICR) instrument used
had limited scan speed, causing long duty cycles. Furthermore, the Chromatographic separation was performed on a Waters
sensitivity of both the ion trap and the FT analyzer (both are tech- (Waters, Milford, MA, USA) BEH C8 column (100 × 1 mm, 1.7 ␮m),
nologies from more than a decade ago) are quite limited when thermostatted to 50 ◦ C in a Dionex Ultimate 3000 RS UHPLC sys-
compared to more timely mass analyzers. tem. Mobile phase A was deionized water containing 1 vol% of 1 M
While most lipidomic research is focused on study of mam- aqueous ammonium formate (final concentration 10 mmol/L) and
malian organisms containing almost exclusively fatty acyls with an 0.1 vol% of formic acid as additives. Mobile Phase B was a mixture of
even number of carbon atoms, lower organisms such as Caenorhab- acetonitrile/isopropanol 5:2 (v/v) with the same additives. Gradient
ditis elegans have a high amount of odd-carbon fatty acyls in their elution started at 50% mobile phase B, rising to 100% B over 40 min;
lipids [6,32–34]. This drastically increases the complexity of the 100% B were held for 10 min and the column was re-equilibrated
lipidome as well as the number of potentially interfering com- with 50% B for 8 min before the next injection. The flow rate was
pounds for each analyte and requires an appropriately powerful 150 ␮L/min, the samples were kept at 8 ◦ C and the injection volume
method for correct analysis. was 2 ␮L.
Here we present a massively improved approach for lipidomic
profiling of even the most complex biological samples with the 2.5. MS method
potential to structurally analyze and quantify hundreds of glycero-
, phospho- and sphingolipids in a single run. Highest selectivity The Orbitrap Velos Pro hybrid mass spectrometer (Thermo
is ensured by combining information from chromatography, high- Fisher Scientific Inc., Waltham, MA, USA) was operated in Data
resolution full scans and tandem mass spectra in conjunction with Dependent Acquisition mode using a HESI II ion source. Prior to
automated data analysis. experiments, lens settings were tuned and source parameters were
optimized to the signal of PE 16:0/18:1 using the protonated and the
deprotonated molecule. Every sample was measured once in pos-
2. Material and methods itive polarity and once in negative polarity. Ion source parameters
for positive polarity were as follows: Source Voltage: 4.5 kV; Source
2.1. Chemicals Temperature: 275 ◦ C; Sheath Gas: 25 arbitrary units; Aux Gas: 9
arbitrary units; Sweep Gas: 0 arbitrary units; Capillary Tempera-
Acetonitrile, isopropanol, methanol, tert-methyl-butyl ether ture: 300 ◦ C. Ion source parameters for negative ion mode were:
(MTBE) (all Chromasolv grade) and ammonium formate (LC/MS Source Voltage: 3.8 kV; Source Temperature: 325 ◦ C; Sheath Gas:
74 A. Triebl et al. / J. Chromatogr. B 1053 (2017) 72–80

30 arbitrary units; Aux Gas: 10 arbitrary units; Sweep Gas: 0 arbi- plementary Table 2 were added to concentrations of 2 ␮mol/L for
trary units; Capillary Temperature: 300 ◦ C. Automatic gain control normalization.
target value was set to 106 ions to enter the mass analyzer, with a The matrix effect was evaluated by comparing the responses of
maximum ion accumulation time of 500 ms. Full scan profile spec- the LIPID MAPS internal standards (listed in Supplementary Table
tra from m/z 400–1200 for positive ion mode and from 400 to 1600 2) analyzed both alone and spiked into a liver lipid extract to con-
in negative ion mode were acquired in the Orbitrap mass analyzer centrations of 2 ␮mol/L.
at a resolution setting of 100 000 at m/z 400. For MS/MS experi-
ments, the 10 most abundant ions of the full scan spectrum were
sequentially fragmented in the ion trap using He as collision gas 3. Results and discussion
(CID, Normalized Collision Energy: 50; Isolation Width: 1.5; Acti-
vation Q: 0.2; Activation Time: 10) and centroided product spectra 3.1. Chromatography
at normal scan rate (33 kDa/s) were collected. The exclusion time
was set to 10 s. We evaluated multiple columns in terms of chromatographic
resolution, peak shape, reproducibility and practicability and found
the BEH C8 column to suit our needs. Supplementary Figs. 1 and 2
2.6. Data processing and quantitation
show example chromatograms, and Supplementary Table 1 shows
performance characteristics for five different RP-HPLC columns
LC/MS data were processed using Lipid Data Analyzer (LDA), as
evaluated. The BEH C8 column provides the necessary chromato-
previously described [35]. Briefly, the algorithm identifies lipids
graphic performance to separate lipids spanning a broad range of
with a 3D algorithm, using the three dimensions m/z, retention
polarity: The very polar species LPC 13:0 and LPC 14:0 are chro-
time, and intensity to correctly integrate peaks, while also taking
matographically separated and both elute well after the solvent
into account the isotopic distribution. MS/MS spectra (displayed,
peak; and there is over 3 min of elution time left after the retention
but not automatically interpreted by the program) were manually
time of TG 60:1 to allow for elution of even longer triacylglycerols.
inspected to obtain information about characteristic head group
Using the same acidic mobile phase for positive and negative
fragments and fatty acyl composition and, if possible, positions, as
ionization we did not observe drastically reduced ionization in neg-
described previously [36–38]. Lipid species contributing less than
ative ion mode but significantly gained selectivity, compared to a
1% to the total amount of the respective lipid class were omitted
formerly used mobile phase without formic acid for analysis in neg-
from the analysis.
ative ion mode. Every lipid elutes at the same retention time both
For quantitation purposes, phosphatidylcholine (PC), phos-
in negative and in positive ion mode, allowing for alignment of
phatidylethanolamine (PE), phosphatidylserine (PS), lysophos-
chromatograms to additionally confirm correct species identifica-
phatidylcholine (LPC), lysophosphatidylethanolamine (LPE) and
tion (Supplementary Figs. 3 and 4). Almost all lipids form sodium
sphingomyelin (SM) were analyzed as [M+H]+ ions, ceramides (Cer)
adducts in positive ion mode and for some lipids (CL and PS), sodium
and hexosylceramides (HexCer) as [M+H−H2 O]+ , and diacylglyc-
adducts even occur in negative ion mode as [M−2H+Na]− [41].
erols (DG) and triacylglycerols (TG) as [M+NH4 ]+ ions in positive
Sodium adducts can be particularly misleading, since they have
polarity, while cardiolipin (CL), phosphatidylinositol (PI) and phos-
only a marginal m/z difference from the protonated form of another
phatidylglycerol (PG) were quantified in negative ion mode as
lipid of the same class. For example, [PC 38:4+Na]+ and [PC 40:7+H]+
[M−H]− ions.
have a difference in molecular mass of only 2.4 mDa which cannot
Lipids were quantified using LIPID MAPS internal standards con-
be baseline separated at the mass resolution used (Supplementary
taining odd-chain fatty acyls for phospholipids or sphingolipids, or
Fig. 4).
deuterated glycerolipids. Quantitation of naturally occurring lipids
Retention time is an essential parameter in lipidomic LC/MS
was performed using a one-point calibration. To 60 ␮L of the liver
data analysis. As previously shown and well-described by the
extract and the A549 cell extract, all LIPID MAPS internal standards
equivalent carbon number model [31,42–44], retention of lipids
listed in chapter 2.2 were added to a concentration of 1.5 ␮M prior
in reversed phase chromatography increases with total number of
to measurement. To 60 ␮L of the C. elegans sample, only PC 25:0,
carbon atoms and decreases with the number of double bonds. The
PE 25:0, PS 25:0, PG 25:0, PI 25:0, the deuterated TG and DG mix,
construction of homologous series for each lipid class can be used
and ceramide/sphingoid internal standard mixture were added at
to discard false positive results from structural isomers belonging
a concentration of 5 ␮M. Lysophospholipids, for which there is no
to different lipid classes, or nominally isobaric interferences such
quantitative LIPID MAPS standard available, were quantified using
as the aforementioned sodium adducts (Fig. 1). Odd-carbon PE and
a 25:0 diacyl species.
PC species, in use as quantitative internal standards, are structural
Lipid shorthand nomenclature is used according to Liebisch et al.
isomers of even-carbon PC and PE, respectively (e.g. internal stan-
[39]. Furthermore, we adhere to the recommendations by D.A.
dard PC 31:1 and analyte PE 34:1), but are clearly distinguishable
Volmer concerning nomenclature in mass spectrometry publica-
by retention time.
tions [40].
The method presented delivers good chromatographic per-
formance for more than 10 phospho-, sphingo- and glycerolipid
2.7. Method validation classes, but analysis of classes such as phosphatidic acid (PA)
and phosphatidylinositolphosphate (PIP) is hindered by poor chro-
Method validation was performed similar to [31]: A liver matographic peak shapes. The strong peak tailing of PA and PIP
lipid extract was spiked with synthetic lipid standards (PC is attributed to interactions of the terminal phosphate group with
12:0/12:0, PE 12:0/12:0, PS 12:0/12:0, PG 12:0/12:0, LPC 15:0/0:0, stainless steel surfaces [45,46], an effect avoided by using phos-
Cer d18:1/17:0, SM d18:1/17:0, TG 17:0/17:0/17:0 and DG phate buffers as HPLC solvent [42], chemical derivatization [47] or
12:0/12:0/0:0) to 13 concentration levels ranging from 1 nM to hydrophilic interaction liquid chromatography [27,48].
100 ␮M to assess linearity (0.001, 0.005, 0.01, 0.05, 0.1, 0.5, 1, 5, 7.5 The duration of a chromatographic run is an important parame-
10, 25, 50, and 100 ␮mol/L). Each concentration level, along with ter influencing total analysis time, and often shorter analysis times
an unspiked lipid extract, was analyzed in triplicate as described in are preferred. However, when analyzing larger sample batches, the
Sections 2.4–2.6. Lipids used for validation purposes were analyzed contribution of chromatographic run time to total analysis time is of
in both polarities (see also Table 1). Internal standards listed in Sup- somewhat lower impact, since sample analysis is completely auto-
A. Triebl et al. / J. Chromatogr. B 1053 (2017) 72–80 75

Table 1
Validation data (unweighted) for lipids in positive and negative mode, determined for one molecular species per lipid class. Values for lower limit of quantitation (LLOQ)
were conservatively determined because of the minimal but detectable natural abundance of the measured analytes. n = 5; n.a.: not applicable (not within linear range).

Accuracy Precision
(%) (RSD%)

Lipid molecular Type of ion LLOQ Linear range R2 Low Medium High Low Medium High
species (fmol on column) (␮M) (0.05 ␮M) (1 ␮M) (50 ␮M) (0.05 ␮M) (1 ␮M) (50 ␮M)

TG 51:0 [M+NH4 ]+ 100 0.05–7.5 0.9874 176 95 n.a. 7.1 1.9 1.6
DG 24:0 [M+NH4 ]+ 100 0.05–10 0.9918 0 1 n.a. 27.1 5.2 4.9
PC 24:0 [M+H]+ 10 0.005–25 0.9981 144 115 n.a. 2.0 1.7 1.9
[M+HCOO]− 10 0.005–25 0.9886 91 113 n.a. 5.4 0.9 2.1
PE 24:0 [M+H]+ 10 0.005–100 0.9957 97 91 59 1.5 2.1 3.2
[M−H]− 10 0.005–10 0.9955 109 121 n.a. 4.1 3.6 2.9
PS 24:0 [M+H]+ 10 0.005–100 0.9981 54 120 82 5.8 1.7 2.1
[M−H]− 10 0.005–100 0.9976 57 126 83 6.8 1.6 1.1
PG 24:0 [M+H]+ 100 0.05–100 0.9982 28 90 102 6.3 1.6 2.5
[M−H]− 10 0.005–100 0.9945 117 122 67 2.6 0.8 1.8
LPC 15:0 [M+H]+ 10 0.005–25 0.9954 124 119 n.a. 2.3 1.6 2.4
[M+HCOO]− 20 0.01–100 0.9939 51 102 71 3.1 1.0 2.0
Cer 17:0 [M+H−H2 O]+ 10 0.005–10 0.9867 78 112 n.a. 4.5 2.8 3.2
[M−H]- 100 0.05–10 0.9940 100 80 n.a. 3.0 2.1 2.1
SM 17:0 [M+H]+ 20 0.01–50 0.9968 135 121 95 1.4 2.4 3.6
[M+HCOO]- 100 0.05–10 0.9940 76 90 n.a. 3.7 1.8 2.6

mated, and dwarfed by the time required for sample preparation is suitable for identification of unexpected or novel lipids, which
particularly data processing. requires no additional chromatographic runs, but only re-analysis
of existing data.
3.2. Lipid detection and identification In contrast to techniques without precursor ion selection such
as MSE , All Ion Fragmentation, All Ions MS/MS or MS/MS ALL, our
The Orbitrap instrument used is able to provide exceptional approach relies on generation of fragment spectra of only one m/z
spectral quality, and routinely reaches values for resolution value. Assignment of fatty acyl composition and position is facil-
(FWHM) of over 110 000 for lysophospholipids, over 90 000 for itated when all fragments displayed ideally belong to only one
phospholipids, over 80 000 for triacylglycerols and over 70 000 for precursor ion. As is the case with all ion trap scans, our MS/MS
cardiolipins. Mass accuracy is usually well below 3 ppm, and below spectra suffer from low mass cutoff. While the major diagnos-
10 ppm even for low-abundance analytes (see also Supplementary tic head group fragment (protonated phosphocholine, m/z 184) of
Tables 7–9). The ability to obtain MS/MS spectra in the ion trap long-chain protonated PC is lost by low mass cutoff, MS/MS spec-
in parallel to full scanning in the Orbitrap is an enormous bene- tra of the corresponding sodium adducts may be interpreted. These
fit. Using this technique, we achieve cycle times of 1.7–2 s (for one show characteristic head group neutral losses of 59 Da (trimethy-
Orbitrap full scan at full resolution and 10 ion trap MS/MS scans), lamine) and 183 Da (phosphocholine) which are not affected by low
which is sufficient to describe almost all analytes with more than mass cutoff. For almost all lipid classes investigated, interpretation
10 data points per mass chromatographic peak. Even without an of both positive and negative MS/MS spectra delivers specific head
inclusion list, for one representative liver sample, tandem mass group fragments as well as fragments indicative of both identities
spectra were available for 127 of the 195 quantified lipid molec- and positions of the fatty acyls. Exceptions are glycerolipids (TGs
ular species, and additionally for all internal standards, resulting and DGs) of which the positions of the fatty acyls cannot easily be
in MS/MS coverage of 65%. This completely untargeted approach

Fig. 1. Construction of homologous series to aid in identification of lipid molecular species. Reversed phase retention behavior of phospholipids is based on number of carbon
atoms and degree of unsaturation. Extracted ion chromatograms of protonated PE 37:4 shows numerous interferences from (a) isobaric PC 34:4, (b) the second isotope of
PE 37:5, and (c) sodiated PE 35:1. Correct peaks identified by Lipid Data Analyzer are filled red, green background denotes that tandem mass spectra of the corresponding
analytes are available. Note that most of the lipid molecular species depicted show multiple chromatographic peaks belonging to structural isomers differing in fatty acid
composition. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
76 A. Triebl et al. / J. Chromatogr. B 1053 (2017) 72–80

structural isomers, and a software for data processing, lipid identi-


fication and quantitation.

3.3. Separation of structural and positional isomers

The chromatographic approach used is able to separate a wide


variety of structural isomers, which can then be identified based
on their retention behavior or tandem mass spectra. The simplest
cases are PC or PE odd-carbon internal standards, which are struc-
tural isomers of even-carbon PE or PC analytes, but can easily be
differentiated. However, many other diacyl phospholipids or tria-
cylglycerols show more than one peak on the lipid species/bond
type level (Fig. 3). Inspection of the tandem mass spectra reveals
that these peaks almost always correspond to structural isomers
with different fatty acyl compositions which are completely or at
Fig. 2. Number of glycerolipids (GL), glycerophospholipids (GP), and sphingolipids least partially chromatographically separated. This is a substantial
(SP) identified in murine liver, A549 cells and C. elegans with a 1% cutoff for each improvement over the method formerly employed in our lab [31].
lipid class. Glycerolipids were not analysed in the A549 sample. Furthermore, it is an advantage over using HILIC, where individ-
ual species within lipid classes are not separated, thus structural
deduced, since the fatty acyls at the sn-1 and sn-3 position behave isomers can also not be chromatographically separated.
identically upon collisional activation. Many lysophospholipids also show double peaks, which are
Using this approach, hundreds of lipids can be identified in a most likely positional isomers (Supplementary Fig. 6). Our obser-
single lipidomic sample. It is necessary to limit oneself concerning vations are consistent with published data [54–57] stating that the
the number of lipid species for several reasons: Firstly, the pareto regioisomers with the acyl group at the sn-2 position elute ear-
principle also applies to lipidomics, and a small number of lipid lier in reversed phase HPLC and upon collisional activation of the
species usually constitutes the bulk of the lipid biomass. An in- protonated molecule show a higher relative intensity of the head
depth analysis of TGs in murine liver shows that out of a total of group-derived fragment (m/z 184 and neutral loss of 141 for LPC
375 identified molecular species, 18 species already represent over and LPE, respectively) as compared to the neutral loss of water.
90%, and 128 species represent over 99.9% of the total class amount However, it is possible that intra-molecular acyl migration, at least
(Supplementary Fig. 5). This effect is even more pronounced for the to some extent, may occur artificially, during sample preparation
class of PI, where only 5 molecular species represent over 95% of the or storage, depending on the exact circumstances [58].
total class amount. Thus it is necessary to introduce a cut-off simply
to reduce the number of molecular species which contribute only 3.4. Quantitation
negligibly to the whole lipid class. Another reason why we do not
advocate analyzing extremely low-abundance species with global One advantage of using reversed phase chromatography is that
methods is that these species tend to have high values of standard lipid species differing in one double bond are baseline separated by
deviation, increasing the chances of finding no difference where in chromatography, so there is no interference in m/z from isotopo-
fact there is one. We instead argue that extremely low-abundance logues containing two 13 C atoms. This is an advantage over shotgun
compounds should be quantified with specialized methods and not or HILIC approaches, where either all analytes, or all lipid species
with global approaches. For these reasons, a class-specific cut-off of one class, respectively, enter the mass analyzer at the same time,
value of 1% was chosen, which means that for each lipid class, all thereby requiring complicated isotope correction or de-isotoping
lipid species which together contribute less than 1% to the total algorithms [4,30,59–61], except when using mass analyzers with
class amount were omitted from the analysis. We argue that this is ultra-high resolving powers [62].
a good compromise between comprehensiveness and data analysis Classical quantitative analysis, as is customary in LC analysis,
effort for a global untargeted approach. relies on calibration curves for each analyte, ideally in an analyte-
In total, 195 lipid species were identified in murine liver (58 free matrix. In LC/MS lipidomics however, this is not possible due
glycerophospholipids, 106 glycerolipids, and 31 sphingolipids), to the lack of analyte-free matrices and the sheer number of ana-
while a total of around 350 lipid species were identified in C. elegans lytes investigated. Stable isotope labeled internal standards, the
(see Fig. 2 for overview and Supplementary Tables 2–4 for complete gold standard of absolute quantitation, are hardly available and
results). The difference in the number of species found is simply due their use is rarely economical. While absolute amounts are often
to the fact that while liver contains in its lipids almost exclusively required, they should be treated with care and knowledge of the
fatty acyls with an even number of carbon atoms, lower organisms limitations of their accuracy. For these reasons, absolute quanti-
such as C. elegans contain a large amount of fatty acyls with an odd tation at the highest level of quality is very difficult to achieve.
number of carbon atoms, thereby drastically increasing the num- Nevertheless, even if absolute values may be subject to error, up-
ber of lipid species. These numbers of lipid species identified are or downregulations of analytes between sample groups are clearly
in accordance with existing lipidomic methods employing a sin- detectable. Therefore we believe that absolute quantification in the
gle LC/MS method [49–51], while analysis of a substantially larger field of LC/MS lipidomics is best considered semi-quantitative and
number of lipids often requires use of multiple analytical methods its value and significance lies in comparing multiple groups (phys-
[52], specialized chromatographic methods, such as supercritical iological/developmental states, wild-type vs. knockout, treatment
fluid chromatography [30], or hyphenated chromatographic meth- vs. control etc.) and focusing on fold changes rather than on abso-
ods, such as on-line or off-line 2D-LC [49,53]. lute amounts, similar to pure profiling methods.
Most importantly however, the presented method includes all The selection of suitable internal standards for lipidomic
necessary steps for large-scale lipidomic measurements for highly analysis is challenging. While quantitative standards containing
complex organisms, such as C. elegans; a sample preparation that odd-chain fatty acyls are used for lipidomic analysis of higher
is applicable to a wide range of biological matrices, a powerful organisms, they are unsuitable when the organism of interest –
chromatographic approach including separation of many fatty acyl such as C. elegans – contains high amounts of odd chain fatty acyls
A. Triebl et al. / J. Chromatogr. B 1053 (2017) 72–80 77

Fig. 3. Separation of PE structural isomers from a C. elegans lipid extract. The chromatogram shows extracted ion chromatograms of deprotonated PE 34:2. Manual inspection
of tandem mass spectra reveals three structural isomers, differing in fatty acyl composition separated by reversed phase chromatography. The positions of the fatty acyls can
be determined by comparing the relative intensities of the carboxylate anions. Tandem mass spectrum of the internal standard PC 12:0/13:0 shows the carboxylates of the
sn-2 fatty acyl of higher relative intensity than of the sn-1 fatty acyl, in accordance with literature data [72].

[63]. A compromise would be the use of internal standards which lysophospholipids are easily distinguished by retention time from
merely correspond to low-abundance endogenous lipid species those generated in the ion source.
and spiking them at relatively high concentrations (e.g. 25:0 phos- Although almost all lipid classes are detectable in both polarities,
pholipid species, as performed here), thereby superimposing the quantitation was only performed for one adduct ion per lipid class.
endogenous concentrations. The C. elegans lipid extract was mea- To increase selectivity, tandem mass spectra of another adduct in
sured with and without addition of the internal standards and the the same polarity, or in the other polarity, can be inspected for
baseline abundance of the analytes corresponding to the internal further confirmation. E.g., while PE was quantified in positive polar-
standards was found to be negligible – either not detected or at least ity (due to the selectivity offered by the neutral loss of the head
90-fold lower than when internal standards were added. 1-(1Z- group), negative polarity is far better suited for identification of
alkenyl)-2-acyl-PC and -PE, also termed plasmalogens, for which the identities and positions of the fatty acyls.
there are no LIPID MAPS quantitative internal standards available,
may be quantified with diacyl–PC and −PE internal standards, as
the responses of PC P-36:1 and PE P-36:1 are within 20% of the
respective diacyl species, investigated in triplicate measurements 3.5. Method validation and evaluation of matrix effect
at concentrations of 0.5 and 5 ␮M for both positive and negative
polarities. We also argue that lysophospholipids may be quantified To preface this chapter, any validation in large-scale lipidomics
using a short-chain diacyl species if no other suitable standard is is a compromise: Ideally, and classically, method validation is
available. performed with one validation standard per analyte in an analyte-
Another point to be considered is that the LIPID MAPS diacyl free matrix. Using hundreds of validation standards is simply not
phospholipids internal standards may contain small amounts of feasible, and analyte-free matrices for lipidomics are simply non-
lysophospholipids, probably derived from chemical degradation existent.
during storage etc. This results in the corresponding lysophospho- The chromatographic setup used is capable of reproducible per-
lipid molecular species being present in an injection containing formance. Retention times of naturally occurring lipids from 13
only LIPID MAPS diacyl phospholipid internal standards. Although lipid classes (TG 52:2, Cer 42:2, HexCer 42:1, CL 72:8, DG 34:1,
their relative abundance may seem negligible (in the case of LPE, LPC 18:0, LPE 18:0, PC 34:1, PE 34:2, PG 34:1, PI 38:4, PE P-38:4,
0.5–2% of the area of the respective diacyl PE species), it should PS 38:4, and SM 42:2) remained constant over the course of 48
be noted that lysophospholipids usually represent only a fraction consecutive injections of a liver extract (average relative standard
of the respective diacyl phospholipid class and therefore, degrada- deviation 0.09%). Autosampler stability over 48 h was assessed by
tion of only a minor amount of diacyl phospholipids could already calculating RSD for the concentrations of these 13 lipid species,
result in an overestimation of the corresponding lysophospho- which were on average 8.6%, indicating no changes in concentra-
lipid species. However, this problem concerns only few molecular tion with prolonged analysis time and storage in the autosampler
species, but their results should be critically evaluated. Source (Supplementary Table 5). Relative standard deviation of the peak
fragmentation in contrast, which would also lead to an artificial area of naturally occuring PC 34:1 in five replicate injections of
increase of lysophospholipids, plays no role here, as the authentic liver, C. elegans and A549 cell lipid extracts was 6.8%, 5.3% and 2.2%,
respectively, and retention time was unaffected by the different
sample matrices (overall relative standard deviation 0.4%).
78 A. Triebl et al. / J. Chromatogr. B 1053 (2017) 72–80

It is well established that the electrospray ionization process has precision is excellent throughout (Table 1), resulting in a mean
a narrow linear dynamic range and the upper concentration limit is relative standard deviation of 7.1% for all analytes quantified.
frequently cited as 10−5 M [64–66], although this value is strongly
expected to depend on the analytes and exact environment. The
4. Conclusion
linearity of the presented method was evaluated for 9 different
lipid classes by spiking a liver extract with a non-endogenous or
A sensitive and highly reliable method for detection and
at least low-abundance lipid species in the range of 1 nM–100 ␮M.
quantitation of molecular species of multiple glycerolipid, glyc-
While some lipids were found to exhibit almost perfect linear-
erophospholipid and sphingolipid classes was developed. The
ity over the whole concentration range investigated (e.g. PG 24:0
method is applicable to a wide variety of biological matrices with a
[M+H]+ , R2 = 0.9989 and [M−H]− R2 = 0.9945), others show a dis-
versatile but simple MTBE extraction procedure. Intact lipid species
tinct saturation-like response at higher concentrations (e.g. PC 24:0
are detected by a high mass resolution Orbitrap Velos Pro hybrid
[M+H]+ ) (Supplementary Fig. 7). It is noteworthy that some lipid
mass spectrometer in both positive and negative polarity, while
classes may have a broader linear range in one polarity than the
tandem mass spectra acquired in the linear ion trap provide deep
other, and also adducts of the same lipid class deviate in their linear
insight into the molecular structure. The reversed phase chro-
range (Table 1).
matography approach used is capable of robust performance, offers
While arguably a better choice as validation standards, the LIPID
important additional selectivity, and is able to resolve structural
MAPS internal standards used for quantitation are supplied only at
and positional isomers. Finally, lipids are identified and quanti-
relatively low concentrations (425 ␮mol/L); thus the lipid species
fied by ample use of quantitative internal standards together with
listed in Chapter 2.7 were chosen as validation standards. The non-
an automated software package, thereby offering a flexible plat-
spiked liver matrix already contains detectable although minute
form for untargeted lipidomic analysis of even the most complex
amounts of the lipid standards added for validation purposes (e.g.
biological samples.
naturally occurring 12:0/12:0 phospholipid species). These were
Method validation indicates a linear range of four orders of mag-
nevertheless chosen for validation because their natural abundance
nitude for most lipid classes investigated, while limits of detection
is insignificant compared to the vast excess of other sample com-
are in the low femtomolar range. The method was applied to murine
ponents and linearity is not negatively affected. While carryover
liver, adenocarcinomic cells, and C. elegans, resulting in quantita-
of the injection system can be ruled out, lower limits of quanti-
tive and structural information about hundreds of lipid species in
tation cannot be classically determined. The limit of quantitation
a manner applicable to large-scale lipidomic studies.
was therefore conservatively determined as the lowest concentra-
tion point which differed substantially from the non-spiked sample
(for an example, see Supplementary Fig. 8). LLOQs vary for different Conflict of interest
lipid classes, but are generally in the range of a few fmol on column
(Table 1). All authors declare no conflict of interest.
While using multiple internal standards for one lipid class is
beneficial, they may differ in their response (i.e. peak area), and Acknowledgements
additionally suffer from matrix effects, a well-known phenomenon
in LC/MS [67–69]. For example, the four PS internal standards differ This work was supported by the Austrian Science Fund (FWF) [P
in their responses by as much as 44% when analyzed at a concentra- 26148].
tion of 1.5 ␮M with and without sample matrix, whereas most other The authors would like to thank Professor Frank Döring
lipid classes analyzed do not exhibit grave matrix effects (Supple- (Christian-Albrechts-Universität zu Kiel, Germany), Dr. Katharina
mentary Table 1). Additionally, instrument response is known to Leithner (Medical University of Graz, Austria), and Dr. Kathrin
depend on factors such as lipid class, acyl chain length and degree Zierler (Karl-Franzens-Universität Graz, Austria) for providing C.
of unsaturation, even within the same lipid class [70,71]. Thus, elegans, A549 cells, and mouse liver samples.
internal standards of differing molecular species and eluting at
different retention times may experience different degrees of ion
suppression, or even ion enhancement, caused by coeluting sample Appendix A. Supplementary data
components. Also, ionization efficiency (i.e. number of ions formed
in the electrospray process) may be different, as solvent composi- Supplementary data associated with this article can be found,
tion is not constant during gradient elution. This effect is observable in the online version, at http://dx.doi.org/10.1016/j.jchromb.2017.
to at least some degree in most lipid classes for which multiple 03.027.
internal standards are available (Supplementary Fig. 9). It is par-
ticularly grave for the class of DG, where the internal standards References
differ enormously with chain length and number of double bonds.
For example, under the conditions employed, the signal of IS d5DG [1] E. Fahy, S. Subramaniam, H.A. Brown, C.K. Glass, A.H. Merrill, R.C. Murphy,
et al., A comprehensive classification system for lipids, J. Lipid Res. 46 (2005)
28:0 is only 2% of the signal of IS d5DG 40:8 even within the same
839–861.
sample. On the other hand, most other lipid classes do not show [2] E. Fahy, S. Subramaniam, R.C. Murphy, M. Nishijima, C.R.H. Raetz, T. Shimizu,
grave differences in instrument response between species differing et al., Update of the LIPID MAPS comprehensive classification system for
in chain length and degree of unsaturation. lipids, J. Lipid Res. 50 (2008) S9–S14.
[3] M.R. Wenk, The emerging field of lipidomics, Nat. Rev. Drug Discov. 4 (2005)
To minimize these problems, the quantitation software 594–610.
employed uses a sophisticated algorithm, explained in greater [4] H.C. Köfeler, A. Fauland, G.N. Rechberger, M. Trötzmüller, Mass spectrometry
detail in the original publication [35], which selects the best inter- based lipidomics: an overview of technological platforms, Metabolites 2
(2012) 19–38.
nal standard, if multiple internal standards are available – as is the [5] C.S. Ejsing, E. Duchoslav, J. Sampaio, K. Simons, R. Bonner, C. Thiele, et al.,
case for most glycerophospholipids. Despite the previously men- Automated identification and quantification of glycerophospholipid
tioned differences in response, this approach results in very good molecular species by multiple precursor ion scanning, Anal. Chem. 78 (2006)
6202–6214.
values for accuracy for almost all lipid classes investigated while [6] D. Schwudke, J. Oegema, L. Burton, E. Entchev, J.T. Hannich, C.S. Ejsing, et al.,
Lipid profiling by multiple precursor and neutral loss scanning driven by the
data-dependent acquisition, Anal. Chem. 78 (2006) 585–595.
A. Triebl et al. / J. Chromatogr. B 1053 (2017) 72–80 79

[7] C.S. Ejsing, J.L. Sampaio, V. Surendranath, E. Duchoslav, K. Ekroos, R.W. [32] V. Matyash, G. Liebisch, T.V. Kurzchalia, A. Shevchenko, D. Schwudke, Lipid
Klemm, et al., Global analysis of the yeast lipidome by quantitative shotgun extraction by methyl-tert-butyl ether for high-throughput lipidomics, J. Lipid
mass spectrometry, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 2136–2141. Res. 49 (2008) 1137–1146.
[8] L. Kalvodova, J.L. Sampaio, S. Cordo, C.S. Ejsing, A. Shevchenko, K. Simons, The [33] B. Jenkins, J. West, A. Koulman, A review of odd-Chain fatty acid metabolism
lipidomes of vesicular stomatitis virus semliki forest virus, and the host and the role of pentadecanoic acid (C15:0) and heptadecanoic acid (C17:0) in
plasma membrane analyzed by quantitative shotgun mass spectrometry, J. health and disease, Molecules 20 (2015) 2425–2444.
Virol. 83 (2009) 7996–8003. [34] J. Prasain, L. Wilson, H. Hoang, R. Moore, M. Miller, Comparative lipidomics of
[9] K. Schuhmann, R. Herzog, D. Schwudke, W. Metelmann-Strupat, S.R. caenorhabditis elegans metabolic disease models by SWATH non-targeted
Bornstein, A. Shevchenko, Bottom-up shotgun lipidomics by higher energy tandem mass spectrometry, Metabolites 5 (2015) 677–696.
collisional dissociation on LTQ Orbitrap mass spectrometers, Anal. Chem. 83 [35] J. Hartler, M. Troetzmueller, C. Chitraju, F. Spener, H.C. Kofeler, G.G. Thallinger,
(2011) 5480–5487. Lipid data analyzer: unattended identification and quantitation of lipids in
[10] D. Schwudke, K. Schuhmann, R. Herzog, S.R. Bornstein, A. Shevchenko, LC-MS data, Bioinformatics 27 (2011) 572–577.
Shotgun lipidomics on high resolution mass spectrometers, Cold Spring Harb. [36] W.J. Griffiths, Tandem mass spectrometry in the study of fatty acids bile acids,
Perspect. Biol. 3 (2011) a004614. and steroids, Mass Spectrom. Rev. 22 (2003) 81–152.
[11] K. Schuhmann, R. Almeida, M. Baumert, R. Herzog, S.R. Bornstein, A. [37] R.C. Murphy, P.F. James, A.M. McAnoy, J. Krank, E. Duchoslav, R.M. Barkley,
Shevchenko, Shotgun lipidomics on a LTQ Orbitrap mass spectrometer by Detection of the abundance of diacylglycerol and triacylglycerol molecular
successive switching between acquisition polarity modes, J. Mass Spectrom. species in cells using neutral loss mass spectrometry, Anal. Biochem. 366
47 (2012) 96–104. (2007) 59–70.
[12] K. Sandra, A. dos, S. Pereira, G. Vanhoenacker, F. David, P. Sandra, [38] F.-F. Hsu, J. Turk, Electrospray ionization with low-energy collisionally
Comprehensive blood plasma lipidomics by liquid activated dissociation tandem mass spectrometry of glycerophospholipids:
chromatography/quadrupole time-of-flight mass spectrometry, J. mechanisms of fragmentation and structural characterization, J. Chromatogr.
Chromatogr. A 1217 (2010) 4087–4099. B. 877 (2009) 2673–2695.
[13] K. Yang, Z. Zhao, R.W. Gross, X. Han, Systematic analysis of choline-containing [39] G. Liebisch, J.A. Vizcaíno, H. Köfeler, M. Trötzmüller, W.J. Griffiths, G. Schmitz,
phospholipids using multi-dimensional mass spectrometry-based shotgun et al., Shorthand notation for lipid structures derived from mass
lipidomics, J. Chromatogr. B 877 (2009) 2924–2936. spectrometry, J. Lipid Res. 54 (2013) 1523–1530.
[14] J. a. Bowden, J.T. Bangma, J.R. Kucklick, Development of an automated [40] D.A. Volmer, Terms and acronyms that should be avoided in mass
multi-injection shotgun lipidomics approach using a triple quadrupole mass spectrometry publications, Rapid Commun. Mass Spectrom. 28 (2014)
spectrometer, Lipids 49 (2014) 609–619. 1853–1854.
[15] C. Wang, M. Wang, X. Han, Comprehensive and quantitative analysis of [41] F.-F. Hsu, J. Turk, Characterization of cardiolipin from Escherichia coli by
lysophospholipid molecular species present in obese mouse liver by shotgun electrospray ionization with multiple stage quadrupole ion-trap mass
lipidomics, Anal. Chem. 87 (2015) 4879–4887. spectrometric analysis of [M−2H+Na]- ions, J. Am. Soc. Mass Spectrom. 17
[16] X. Han, K. Yang, J. Yang, K.N. Fikes, H. Cheng, R.W. Gross, Factors influencing (2006) 420–429.
the electrospray intrasource separation and selective ionization of [42] O.L. Knittelfelder, B.P. Weberhofer, T.O. Eichmann, S.D. Kohlwein, G.N.
glycerophospholipids, J. Am. Soc. Mass Spectrom. 17 (2006) 264–274. Rechberger, A versatile ultra-high performance LC-MS method for lipid
[17] M. Kliman, J.C. May, J. a McLean, Lipid analysis and lipidomics by structurally profiling, J. Chromatogr. B. 951–952 (2014) 119–128.
selective ion mobility-mass spectrometry, Biochim. Biophys. Acta – Mol. Cell [43] M. Holčapek, M. Ovčačíková, M. Lísa, E. Cífková, T. Hájek, Continuous
Biol. Lipids 1811 (2011) 935–945. comprehensive two-dimensional liquid chromatography-electrospray
[18] A. a Shvartsburg, G. Isaac, N. Leveque, R.D. Smith, T.O. Metz, Separation and ionization mass spectrometry of complex lipidomic samples, Anal. Bioanal.
classification of lipids using differential ion mobility spectrometry, J. Am. Soc. Chem. 407 (2015) 5033–5043.
Mass Spectrom. 22 (2011) 1146–1155. [44] A. Triebl, S. Weissengruber, M. Trötzmüller, E. Lankmayr, H. Köfeler,
[19] P.R.S. Baker, A.M. Armando, J.L. Campbell, O. Quehenberger, E.a. Dennis, Quantitative analysis of N-acylphosphatidylethanolamine molecular species
Three-dimensional enhanced lipidomics analysis combining UPLC, in rat brain using solid-phase extraction combined with reversed-phase
differential ion mobility spectrometry, and mass spectrometric separation chromatography and tandem mass spectrometry, J. Sep. Sci. 39 (2016)
strategies, J. Lipid Res 55 (2014) 2432–2442. 2474–2480.
[20] T.P.I. Lintonen, P.R.S. Baker, M. Suoniemi, B.K. Ubhi, K.M. Koistinen, E. [45] A. Wakamatsu, K. Morimoto, M. Shimizu, S. Kudoh, A severe peak tailing of
Duchoslav, et al., Differential mobility spectrometry-driven shotgun phosphate compounds caused by interaction with stainless steel used for
lipidomics, Anal. Chem. 86 (2014) 9662–9669. liquid chromatography and electrospray mass spectrometry, J. Sep. Sci. 28
[21] A.T. Maccarone, J. Duldig, T.W. Mitchell, S.J. Blanksby, E. Duchoslav, J.L. (2005) 1823–1830.
Campbell, Characterization of acyl chain position in unsaturated [46] Y. Asakawa, N. Tokida, C. Ozawa, M. Ishiba, O. Tagaya, N. Asakawa,
phosphatidylcholines using differential mobility-mass spectrometry, J. Lipid Suppression effects of carbonate on the interaction between stainless steel
Res. 55 (2014) 1668–1677. and phosphate groups of phosphate compounds in high-performance liquid
[22] M. Groessl, S. Graf, R. Knochenmuss, High resolution ion mobility-mass chromatography and electrospray ionization mass spectrometry, J.
spectrometry for separation and identification of isomeric lipids, Analyst 140 Chromatogr. A 1198–1199 (2008) 80–86.
(2015) 6904–6911. [47] M.J.O. Wakelam, The uses and limitations of the analysis of cellular
[23] J. a. Hankin, R.C. Murphy, R.M. Barkley, M.a. Gijón, Ion mobility and tandem phosphoinositides by lipidomic and imaging methodologies, Biochim.
mass spectrometry of phosphatidylglycerol and bis(monoacylglycerol) Biophys. Acta – Mol. Cell Biol. Lipids 1841 (2014) 1102–1107.
phosphate (BMP), Int. J. Mass Spectrom. 378 (2015) 255–263. [48] E. Cífková, R. Hájek, M. Lísa, M. Holčapek, Hydrophilic interaction liquid
[24] R. Herzog, D. Schwudke, K. Schuhmann, J.L. Sampaio, S.R. Bornstein, M. chromatography-mass spectrometry of (lyso)phosphatidic acids,
Schroeder, et al., A novel informatics concept for high-throughput shotgun (lyso)phosphatidylserines and other lipid classes, J. Chromatogr. A 1439
lipidomics based on the molecular fragmentation query language, Genome (2016) 65–73.
Biol. 12 (2011) R8. [49] D.Y. Bang, M.H. Moon, On-line two-dimensional capillary strong anion
[25] F.-F. Hsu, J. Turk, Studies on phosphatidylserine by tandem quadrupole and exchange/reversed phase liquid chromatography–tandem mass spectrometry
multiple stage quadrupole ion-trap mass spectrometry with electrospray for comprehensive lipid analysis, J. Chromatogr. A 1310 (2013) 82–90.
ionization: structural characterization and the fragmentation processes, J. [50] M. Sarafian, M.R. Gaudin, F. Lewis, E. Martin, et al., Objective set of criteria for
Am. Soc. Mass Spectrom. 16 (2005) 1510–1522. optimization of sample preparation procedures for ultra-high throughput
[26] J.G. Bollinger, H. Ii, M. Sadilek, M.H. Gelb, Improved method for the untargeted blood plasma lipid profiling by ultra performance liquid
quantification of lysophospholipids including enol ether species by liquid chromatography–mass spectrometry, Anal. Chem. 86 (2014) 5766–5774.
chromatography-tandem mass spectrometry, J. Lipid Res. 51 (2010) 440–447. [51] M. Ovčačíková, M. Lísa, E. Cífková, M. Holčapek, Retention behavior of lipids in
[27] A. Triebl, M. Trötzmüller, A. Eberl, P. Hanel, J. Hartler, H.C. Köfeler, reversed-phase ultrahigh-performance liquid chromatography–electrospray
Quantitation of phosphatidic acid and lysophosphatidic acid molecular ionization mass spectrometry, J. Chromatogr. A 1450 (2016) 76–85.
species using hydrophilic interaction liquid chromatography coupled to [52] O. Quehenberger, A.M. Armando, A.H. Brown, S.B. Milne, D.S. Myers, A.H.
electrospray ionization high resolution mass spectrometry, J. Chromatogr. A Merrill, et al., Lipidomics reveals a remarkable diversity of lipids in human
1347 (2014) 104–110. plasma, J. Lipid Res. 51 (2010) 3299–3305.
[28] M. Holčapek, R. Jirásko, M. Lísa, Recent developments in liquid [53] M. Lísa, E. Cífková, M. Holčapek, Lipidomic profiling of biological tissues using
chromatography-mass spectrometry and related techniques, J. Chromatogr. A off-line two-dimensional high-performance liquid chromatography-mass
1259 (2012) 3–15. spectrometry, J. Chromatogr. A 1218 (2011) 5146–5156.
[29] T. Bamba, J.W. Lee, A. Matsubara, E. Fukusaki, Metabolic profiling of lipids by [54] M.H. Creer, R.W. Gross, Separation of isomeric lysophospholipids by reverse
supercritical fluid chromatography/mass spectrometry, J. Chromatogr. A 1250 phase HPLC, Lipids 20 (1985) 922–928.
(2012) 212–219. [55] M. Murph, T. Tanaka, J. Pang, E. Felix, S. Liu, R. Trost, et al., Liquid
[30] M. Lísa, M. Holčapek, High-throughput and comprehensive lipidomic analysis chromatography mass spectrometry for quantifying plasma
using ultrahigh-Performance supercritical fluid chromatography–mass lysophospholipids: potential biomarkers for cancer diagnosis, Methods
spectrometry, Anal. Chem. 87 (2015) 7187–7195. Enzymol. (2007) 1–25.
[31] A. Fauland, H. Köfeler, M. Trötzmüller, A. Knopf, J. Hartler, A. Eberl, et al., A [56] J.Y. Lee, H.K. Min, M.H. Moon, Simultaneous profiling of lysophospholipids
comprehensive method for lipid profiling by liquid chromatography-ion and phospholipids from human plasma by nanoflow liquid
cyclotron resonance mass spectrometry, J. Lipid Res. 52 (2011) 2314–2322. chromatography-tandem mass spectrometry, Anal. Bioanal. Chem. 400 (2011)
2953–2961.
80 A. Triebl et al. / J. Chromatogr. B 1053 (2017) 72–80

[57] K.M. Koistinen, M. Suoniemi, H. Simolin, K. Ekroos, Quantitative [65] D.R. Zook, A.P. Bruins, On cluster ions, ion transmission, and linear dynamic
lysophospholipidomics in human plasma and skin by LC–MS/MS, Anal. range limitations in electrospray (ionspray) mass spectrometry, Int. J. Mass
Bioanal. Chem. 407 (2015) 5091–5099. Spectrom. Ion Process. 162 (1997) 129–147.
[58] M. Okudaira, A. Inoue, A. Shuto, K. Nakanaga, K. Kano, K. Makide, et al., [66] N.B. Cech, C.G. Enke, Practical implications of some recent studies in
Separation and quantification of 2-acyl-1-lysophospholipids and electrospray ionization fundamentals, Mass Spectrom. Rev. 20 (2001)
1-acyl-2-lysophospholipids in biological samples by LC-MS/MS, J. Lipid Res. 362–387.
55 (2014) 2178–2192. [67] B.K. Matuszewski, M.L. Constanzer, C.M. Chavez-Eng, Strategies for the
[59] G. Liebisch, B. Lieser, J. Rathenberg, W. Drobnik, G. Schmitz, High-throughput assessment of matrix effect in quantitative bioanalytical methods based on
quantification of phosphatidylcholine and sphingomyelin by electrospray HPLC-MS/MS, Anal. Chem. 75 (2003) 3019–3030.
ionization tandem mass spectrometry coupled with isotope correction [68] P.J. Taylor, Matrix effects: the Achilles heel of quantitative high-performance
algorithm, Biochim. Biophys. Acta − Mol. Cell Biol. Lipids 1686 (2004) liquid chromatography–electrospray–tandem mass spectrometry, Clin.
108–117. Biochem. 38 (2005) 328–334.
[60] T. Hyötyläinen, M. Orešič, Systems biology strategies to study lipidomes in [69] H. Trufelli, P. Palma, G. Famiglini, A. Cappiello, An overview of matrix effects
health and disease, Prog. Lipid Res. 55 (2014) 43–60. in liquid chromatography-mass spectrometry, Mass Spectrom. Rev. 30 (2011)
[61] X. Han, K. Yang, R.W. Gross, Multi-dimensional mass spectrometry-based 491–509.
shotgun lipidomics and novel strategies for lipidomic analyses, Mass [70] M. Koivusalo, P. Haimi, L. Heikinheimo, R. Kostiainen, P. Somerharju,
Spectrom. Rev. 31 (2012) 134–178. Quantitative determination of phospholipid compositions by ESI–MS: effects
[62] R. Almeida, J.K. Pauling, E. Sokol, H.K. Hannibal-Bach, C.S. Ejsing, of acyl chain length, unsaturation, and lipid concentration on instrument
Comprehensive lipidome analysis by shotgun lipidomics on a hybrid response, J. Lipid Res. 42 (2001) 663–672.
quadrupole-orbitrap-linear ion trap mass spectrometer, J. Am. Soc. Mass [71] S. Khoury, N. El Banna, S. Tfaili, P. Chaminade, A study of inter-species ion
Spectrom. 26 (2015) 133–148. suppression in electrospray ionization-mass spectrometry of some
[63] T.V. Witting, S. Maier, Optimizing a ultrahigh pressure liquid phospholipid classes, Anal. Bioanal. Chem. 408 (2016) 1453–1465.
chromatography-time of flight-mass spectrometry approach using a novel [72] F.F. Hsu, J. Turk, Charge-remote and charge-driven fragmentation processes in
sub-2 ␮m core–shell particle for in depth lipidomic profiling of diacyl glycerophosphoethanolamine upon low-energy collisional activation: a
Caenorhabditis elegans, J. Chromatogr. A 1359 (2014) 91–99. mechanistic proposal, J. Am. Soc. Mass Spectrom. 11 (2000) 892–899.
[64] K. Tang, J.S. Page, R.D. Smith, Charge competition and the linear dynamic
range of detection in electrospray ionization mass spectrometry, J. Am. Soc.
Mass Spectrom. 15 (2004) 1416–1423.

You might also like