• No results found

University of Groningen Applying lipidomics strategies to study lipid metabolic diseases Zhang, Wenxuan

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Applying lipidomics strategies to study lipid metabolic diseases Zhang, Wenxuan"

Copied!
29
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Applying lipidomics strategies to study lipid metabolic diseases

Zhang, Wenxuan

DOI:

10.33612/diss.169407826

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2021

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Zhang, W. (2021). Applying lipidomics strategies to study lipid metabolic diseases. University of Groningen. https://doi.org/10.33612/diss.169407826

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 2

Omics technology: lipidomics and its

pitfalls during the preanalytical stage

Andres Gil1†, Wenxuan Zhang2†, Justina C. Wolters2,  Hjalmar Permentier3,

Peter Horvatovich1,M. Rebecca Heiner-Fokkema4, Dirk-Jan Reijngoud2, Rainer

Bischoff1*

These authors contributed equally to this work

1Department of Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.2 Systems Medicine and Metabolic Signaling, Department of Pediatrics, University Medical Center Groningen, University of Groningen, PO Box 30.001 9700RB Groningen, The Netherlands. 3Interfaculty Mass Spectrometry Center, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.4Laboratory of Metabolic Disease, Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, PO Box 30.001 9700RB Groningen, The Netherlands

Encyclopedia of Analytical Science. Elsevier; 2019:70-81. doi:10.1016/B978-0-12-409547-2.14002-8

(3)

Introduction

Systems biology is an evolving multidisciplinary field that attempts to provide a collective insight into complex and dynamic biological processes by understanding and integrating the knowledge of biochemical networks at all levels, from molecules to entire organisms. In this regard, an important milestone during the last decades has been the development of comprehensive methods or so called “omic” technologies (genomics, transcriptomics, proteomics and metabolomics), that ideally allow to monitor and characterize all members of families of cellular molecules including DNA/RNA, proteins and metabolites in a single analysis.

Metabolomics is the most recent member of the omic technologies and in comparison to genomics and proteomics, which focus on gene expression and protein products, it focuses on the downstream output of biochemical networks within cells. Thus, metabolomics is considered to reveal the last part of the “omics spectrum” and therefore to be a close representation of a cellular or organismal phenotype. Lipids are a subset of the metabolome representing 70% of the entries in the Human Metabolome Database (HMDB)1. It is thus no surprise that

lipidomics is considered a separate discipline besides metabolomics (Figure 1). Numerous studies have shown that lipids have important signaling functions in addition to their role in cellular membranes and the provision of energy. Lipid-mediated signaling may be altered in widespread human diseases, including cardiovascular disease, diabetes type 2, Alzheimer’s disease, inborn errors of metabolism and cancer1–3. This has consequently accelerated the development

of the lipidomics field mainly focusing on the identification of biomarkers for diagnostic purposes4,5.

Lipidomics

The term lipidomics, introduced in 2003 by Spener et al.6 and Han & Gross7,

refers to the identification and quantitation of the entire collection of chemically distinct lipid species that collectively comprise the lipidome in a cell, tissue, or biological fluid.

Usually lipids produced in an organism, are viewed merely as intermediates and/or end points of biosynthesis. Nonetheless, it is becoming clearer that gaining a detailed view of the lipidome (identity, concentration, biosynthesis and function) is important for our understanding of the functioning of cells and organisms within their respective environment. As such, this knowledge forms an integral part of systems biology. Lipids can be studied with two approaches,

(4)

2

untargeted lipidomics and targeted lipid analysis8,9, each with their own

strengths and limitations.

Untargeted lipidomics versus targeted lipid analysis

Untargeted or global lipidomics is a hypothesis-free approach that aims at comprehensively measuring levels of all lipid species in a sample, including lipids with structures that have yet to be elucidated. The objective of performing a hypothesis-free analysis is acquiring data with the widest possible coverage of the lipidome followed by identification of those lipids whose relative concentrations change following a perturbation (e.g., a gene knockout, a change in nutrients, physical exercise). Due to its holistic nature (measurement of both known and unidentified lipids), internal standards are not typically used, thus complicating the quantitative determination of absolute lipid levels and making it more useful for the discovery of associations between components of the lipidome.

Figure 1. The human metabolic Network. Modified from the KEGG Pathway Database

KEGG PATHWAY: Metabolic pathways—Homo sapiens (human) http://www.genome.jp/ kegg-bin/show_pathway?scale1⁄41.0&query1⁄4&map1⁄4hsa01100&scale1⁄40.35&auto_ image1⁄4&show_description1⁄4hide&multi_query1⁄4&show_module_list1⁄4 (accessed Nov 14, 2017).

(5)

Targeted lipid analysis, on the other hand, has the goal of quantifying selected lipids that are of specific biological interest, for example as part of a biochemical pathway or network (e.g., prostaglandin synthesis). By using internal standards (usually isotopically labeled lipids), absolute concentrations or amounts can be determined. Targeted lipid analysis requires a thorough understanding of the metabolic pathway or network under study in order to define the lipid species that shall be monitored. In contrast to untargeted lipidomics, sample preparation can be tailored to enrich the lipids of interest while removing highly abundant interfering species. In addition, since all analytes are defined from the very beginning, analytical artifacts are not carried through to the downstream data analysis10. Thus, absolute concentrations of lipids in the sample can be

determined with high accuracy, precision, sensitivity, and specificity (Figure 2). Since the targeted analysis offers a quantitative picture of selected compounds in a specific part of the lipidome, it is often used as a complementary tool to untargeted lipidomics, notably to follow up on a discovery lipidomics study. The general preanalytical steps for both, untargeted lipidomics and targeted lipid analysis, are rather similar (Figure 3), while data analysis differs significantly. However, there is a lack of awareness on how lipid stability is affected during sample preparation. Thus, below we focus on common pitfalls in a typical lipid analysis (untargeted and targeted) workflows and suggest ways to avoid them.

Lipid stability

The stability of lipid molecules, that is, their susceptibility to chemical degradation during sample handling, is highly related to their structural features. Ester bonds such as those linking fatty acid groups in glycerophospholipids are susceptible to hydrolysis11. Saturated fatty acids groups (e.g., myristic acid) are

rather stable, While lipids containing unsaturated fatty acids, with one or more double bonds (e.g., dioleic acid), are susceptible to oxidation in the presence of air (oxygen), which is enhanced by exposure to light and the presence of heavy metal ions (notably Fe2+)11.

Temperature and pH are critical variables during the preanalytical phase for any lipid analysis. Temperatures between 4 and 25°C and pH values ranging from 2 to 8 are typical conditions during lipid analysis. Moreover, chemical environments rich in water and oxygen, the presence of light and sample handling times up to 24 h are also common in lipid analysis. Not surprisingly, all of these conditions might be associated with chemical degradation processes affecting lipid species present in almost any sample matrix, as well as in plain standard solutions. Although there are guidelines for dealing with stability as a key issue in the

(6)

2

validation of bioanalytical methods, these usually focus on single analytes12.

Particularly for untargeted lipidomics studies, sample stability is far more difficult to assess, given the complexity of the samples that are composed of hundreds to tens of thousands of analytes of varying chemical composition and stability. Some studies tried to tackle this problem for proteomics and metabolomics but there is no consensus on how to approach stability issues for complex ‘Omics’ samples13,14.

Figure 2. Differences and similarities between untargeted lipidomics and targeted lipid analysis.

The stability of lipidomes under different conditions has not been assessed in great detail despite 20 years of development in the field15. The main reason

for this is due to limitations in throughput and the lack of suitable reference materials, since it is not possible to follow the typical strategy of targeted lipid analysis in which isotopically labeled internal (homologue) standards are used for each analyte under study. A strategy to overcome this issue might be the use of standardized reference lipidomes for comparison purposes. However, stability assessment is mandatory in lipidomics to produce reliable and reproducible results. In this sense, general considerations must be taken

(7)

into account in order to minimize the breakdown of lipids species across the workflow, as discussed below.

The lipidomics workflow and possible pitfalls

Lipidomics is increasingly used in numerous research fields, including food and nutritional16,17, clinical18,19, biological20, pharmaceutical21 and biotechnological22

disciplines. Next to high-quality analytical instrumentation and well-trained laboratory staff, it is critical to have experienced personnel in data processing and statistical data analysis as fundamentals to the success of lipidomics projects. Since lipidomics aims to be a comprehensive methodology, all manipulations of the sample from the preanalytical phase to the analysis of the data will have an effect on the final result. Consequently, standardization of procedures is a prerequisite for obtaining reliable results.

Figure 3. Elements of a general experimental design and workflow for lipid analysis (untargeted

and targeted).

Since lipidomics is a field of research derived from metabolomics, it basically follows the same experimental design and workflow. As shown in Figure 3, the preanalytical phase of a typical lipidomics/metabolomics research project consists of two major steps. First, the phase of defining the biological/ clinical question and the corresponding experimental design, and second, the bioanalytical phase during which sampling, storage, sample handling, metabolism quenching, lipid/metabolite extraction and LC-MS protocols need to be defined and executed. These processes must be strictly controlled

(8)

2

and performed according to standardized procedures, to avoid undesired alterations of the samples that may lead to biased results.

Experimental design

Lipidomics in body fluids

One of the critical steps in lipidomics, as in any other ‘Omics’ approach, is the selection of appropriate samples to address the biological question. Currently, for clinical lipidomics research, tissue or biofluid samples are used23. Due

to the ease of collection, plasma, serum and urine have been widely used for the prognosis or diagnosis of many diseases. Additionally, blood (serum and plasma) and urine are integrative biofluids that reflect changes in the (patho) physiology of the entire organism in a single sample24, which may, however, be

problematic, as localized changes due to early disease development may be difficult to detect.

Serum is obtained by allowing the natural process of clotting to occur in whole blood at room temperature (possibly with the addition of clotting activators), followed by centrifugation to isolate the supernatant, while plasma is produced by the removal of cells in a centrifuge after treatment of the blood sample with an anticoagulant to prevent clot formation24,25. Although blood is a common

biofluid used for biomarker discovery, the degree of natural variation for many lipids remains poorly understood, thus complicating the interpretation of whether observed lipid fluctuations are due to (patho)physiological changes related to a disease, therapeutic interventions or other effects such as nutrition or genetic factors. Few attempts have been made to characterize differences in the lipidome between serum and plasma. Ishikawa et al.26 explored matrix-,

age-, gender and stability-associated differences in the lipid profile of plasma versus serum. They found that the clotting process influences the serum lipid profile, since lysophosphatidylcholines (lysoPC) and diacylglycerols (DAG) showed increased levels in comparison to plasma samples. Many sphingomyelin species were significantly higher in females than in males indicating an important gender-associated difference that was independent of age and matrix (plasma and serum). Age-associated differences were more prominent in females than in males in both matrices. Levels of many triacylglycerols were significantly higher in elderly females than in young females. Finally, the authors also demonstrated the negative effect of freeze-thawing samples on plasma and serum lipidomes, as most lipids showed reduced concentrations after freeze-thaw cycles. Even when assessing within-subject variance of the plasma lipidome, Begum et al.27 found important changes related to the effect

(9)

variability of the concentration of lipid species that might be used as baseline values to better identify clinically relevant disease markers.

Urine contains much lower biomolecule concentrations than plasma or serum, notably less protein. Urine contains low-molecular-weight metabolites at rather low concentrations24 but often with considerable biological variability.

Fernández-Peralbo et al.28 proposed that the human urinary metabolome is

potentially more complex than serum and plasma if all potential endogenous metabolites and exogenous compounds originating from the intake of nutrition,

the gut microbiome and the environment are considered28. Lipidomics has

been used as a strategy to discover biomarkers for disease diagnosis in urine. Byeon et al.29 explored the lipidome of patients with Gaucher disease (GD) by

evaluating the difference between urine and plasma after β-glucocerebrosidase enzyme replacement therapy. They found that in both plasma and urine, all the measured monohexosylceramide (MHC) lipid species tended to be increased in patients when compared to healthy controls. More important was the fact that the concentration of these lipid species decreased after receiving the therapy, implicating that MHC lipid species might be excellent candidates in developing an early diagnostic test for GD in both plasma and urine. Despite this successful example, the metabolic homeostasis of the lipidome in urine, plasma or serum is far from established, as little work has focused on understanding the natural variation of the lipidome in humans over time.

Lipidomics in cellular models

In vitro lipidomics studies in cellular models offer some advantages over human or animal studies, including easily controlled experimental variables, greater reproducibility and lower cost. Factors such as age, gender, between-subject variation, and population control baseline values are not relevant when it comes to in vitro studies in cell cultures30. Instead, issues with culture conditions, the

number of replicates and most importantly the difficulty of translating results to the in vivo situation are among the factors that need to be considered for in vitro studies in cellular model systems. Cells need to be cultured prior to any sample preparation procedure and culture conditions can have an effect on the cellular lipidome. To avoid variation, cells need to grow under standardized and controlled conditions. Seeding density, the culture medium, incubation conditions, harvesting and washing steps, are the most important variables that must remain constant. Later, during the collection of cells, extensive washing steps are typically required to avoid remnants of culture medium and buffers that might affect the lipidome. There is currently a lack of information on how these variables might affect cellular lipidomes. Nonetheless, lipidomics has

(10)

2

been used extensively for understanding/interpreting biological responses to diverse stimuli in cell culture systems.

Escherichia coli has been studied extensively due to its importance as a human

pathogen. It is among the best-understood organisms and has served as a valuable model for the study of numerous biological processes, including cellular metabolism31. Saccharomyces cerevisiae (baker’s yeast) is another

widely studied model in lipidomics research, since it is a simple unicellular eukaryotic organism, containing approximately 6000 genes most of which have orthologues (genes in different species that evolved from a common ancestral gene by speciation) in humans. Taking the high conservation of biological processes and cellular structures throughout eukaryotic systems into account32,

it is possible to gain insight into the human lipidome by using yeast as a model. Mammalian cells represent another crucial target of lipidomic studies. Human fibroblasts, human breast cancer cell lines, Madin–Darby canine kidney (MDCK) cells, hepatocytes and Chinese hamster ovary (CHO) cells have all been used to gain an overview of the lipidome in eukaryotic cells. Despite the central role of lipids in cell biology, efforts to systematically define changes of the lipidome in mammalian cells in response to normal cellular function or perturbations are rarely found33. While there is considerable knowledge about the biochemical

pathways that lead to the synthesis and degradation of the major lipid classes, there is comparatively little research on the interactions between these

pathways under normal and pathophysiological conditions. Dennis et al.33

characterized changes in the lipidome of macrophages in response to Kdo2-lipid A, a highly specific Toll-Like Receptor ligand that mimics bacterial infection. By quantitatively monitoring the changes, they discovered lipid species not previously reported in macrophages including dihomoprostaglandins, ether-linked triglycerides, ether-linked phosphatidylinositol, ether-linked phosphatidylserine, phosphatidylthreonine, N-acyl phosphatidylserine, and deoxyceramides. This illustrates the potential of lipidomics as a discovery tool.

Bioanalysis

Sample processing

As discussed above, a meticulous study design and careful optimization of pre-analytical steps are vital to ensure the quality of the data generated by the subsequent analyses. In lipidomics, the bioanalysis phase is divided into 4 major steps comprising sample collection (typically blood and/or urine), sample storage, sample handling and lipid extraction (Figure 3).

(11)

Sample collection

Probably the most important part of any large-scale lipidomics study is collection of the sample set. The sampling phase is frequently embedded in the clinical workflow and therefore is one of the less standardized processes. Thus, whereas a failed LC-MS analysis can be repeated or excluded in extreme cases, inconsistencies during sample collection may go unnoticed and invalidate the entire study24,34. The impact of devices used for obtaining and storing the samples

on the analysis is often overlooked. They can be a source of contaminants, particularly polymers such as polyethylene glycols and phthalates, which may interfere with the subsequent analyses and affect the results24.

Besides the storage devices, the sampling procedure by itself may affect the result. Preparation of serum requires activation of the blood clotting process, during which a range of enzymes is activated and which may take 30-60 min at room temperature increasing the risk of enzymatic conversions and other degradation processes25,35. In order to avoid this as much as possible, it is

recommended to clot the blood at reduced temperatures (4°C), although enzymatic activity is not completely inhibited until temperatures below −56°C are reached24.

Plasma preparation from blood, on the other hand, requires the use of anticoagulants such as heparin, sodium citrate or ethylenediaminetetraacetic acid (EDTA) followed by a centrifugation step at 4°C to separate plasma from the other blood components including red and white blood cells as well as platelets. Platelet activation is associated with significant changes to membrane lipids, and formation of diverse bioactive lipids that play essential roles in hemostasis. Therefore, the use of procedures to prepare platelet-rich and platelet-poor plasma samples must be considered when performing lipid analysis36. Heparin

is a known platelet activator that may have an important influence on changes

in the lipidome of plasma samples37. However, anticoagulants may introduce

analytical bias and their effect on the lipidome should be evaluated before large-scale sample collection and biobanking35. Gonzales-Covarrubias et al., found

that the effect of citrate, EDTA, and heparin on solution pH or ionic strength may have an important effect on decreasing or increasing the LC-MS response of lipids38. They argue, for example, that during the extraction procedure a high

ionic strength might increase protein denaturation and consequently increase the release of lipids from lipoproteins. Furthermore, the molecular weights of citrate and EDTA are in the same range as endogenous low molecular weight fatty acids (FA), so these anticoagulants might affect the analysis, if targeting this type of FA is the purpose of the study. Lipids tend to form adducts with sodium

(12)

2

and potassium ions in the absence of chelating agents leading to ionization

suppression effects upon electrospray ionization mass spectrometry39.

Gonzales-Covarrubias et al. suggest that some of the observed differences in LC-MS signals in relation to the use anticoagulants might be due to the presence of lithium, as heparin is usually as a lithium salt. This is in agreement with the observation of other researchers that have shown that the presence of lithium-heparin affects the LC–MS response of small molecules differentially38,40.

Hemolysis during sampling is another possible caveat that may affect the overall lipid profile. Previously, Yin et al.34 showed that approximately 18 % of the

detected mass signals in a non-targeted metabolomics experiment are affected by hemolysis. Hemolytic samples usually contain increased concentrations of lipids mainly originating from the cellular membranes of erythrocytes41.

Moreover, the release of intracellular components during the hemolytic process is another major issue that must be avoided, since active enzymes, that might significantly alter the lipidome, may also be released.

For urine analysis, the sampling procedure and sampling time can vary significantly. To evaluate the global status of an individual’s lipidome, it is preferable to use 24 h urine sampling to avoid the large variation in lipid profiles with shorter collection periods28. Furthermore, treating urine samples

with additives to increase lipid solubility and/or to minimize interaction with container surfaces is essential. As an example, Silvester et al.42 established that

by adding surfactants, urine metabolites with lipophilic characteristics could be quantified without losses due to unspecific adsorptive processes to the surface of containers, in which the samples were collected, stored or processed. Surfactants may, however, affect the subsequent LC-MS analysis leading to ionization suppression and interfering signals as well as to accumulation on the LC column. The addition of preservatives, such as boric acid or sodium azide, is a common procedure for urine samples to prevent bacterial growth during storage43. However, the effect of such reagents on the urine lipidome has not yet

been addressed.

For cellular models, sampling involves separating the cellular material from the cell culture medium, and usually a subsequent quenching process to avoid enzymatic degradation of lipid species (discussed below). For mammalian cells, there are some special considerations depending on whether one uses adherent or non-adherent cells. For non-adherent cells, the pellet can be obtained by centrifugation or filtration. For adherent cells, trypsin, EDTA or scrapers are required to detach the cells from the surface of the culture dish44. The influence

(13)

of this process on lipid profiles has not been addressed. Furthermore, culture flasks are usually made of plastic materials that can introduce contaminants, which may affect lipid analysis.

Sample storage

Storage is another important step of the preanalytical phase, as sample content may be altered during storage. Irrespective of the type of sample, be it biofluids, tissues or cells, storage should be done at the lowest temperature available, typically at -80°C, prior to any further manipulation of the samples45. This helps

to inhibit enzymatic activity, and to reduce the rate of oxidation and hydrolytic degradation of lipids containing unsaturated fatty acids. It is furthermore recommended to minimize the number of freeze–thaw cycles and to rapidly freeze (ideally using liquid nitrogen) and store pre-aliquoted samples to decrease the potential for degradation as much as possible25,28. Particularly for

cells, controlled freezing in the presence of cryoprotective solvents must be done in order to maintain the integrity of the cellular membrane, thus avoiding changes in the intracellular lipidome45.

Quenching of metabolism

Lipid stability may be affected by chemical processes as well as by enzymatic degradation. Notably remaining enzymatic activity in the sample may alter the lipidome during storage and/or sample preparation, for example during freeze-thaw cycles25. Cellular metabolism adjusts rapidly to changes in the environment.

Since lipids are coupled through metabolic networks, these changes can quickly generate important alterations in the lipid profiles46. Therefore, another

important step during the lipidomics workflow, particularly important for cell and tissue samples, is quenching of enzymatic activity. Quenching aims to stop metabolism and prevent the turnover of lipids, maintaining their concentrations at the original level. Rapid quenching of metabolism is usually achieved through fast changes in temperature or pH47.

Quenching procedures were originally developed for and applied to yeast, but have later also been used in studies of different bacteria, mammalian cells,

tissues and biofluids44,48. Quenching may be achieved by a rapid change of

temperature to either low (e.g. below -40°C) or high temperatures (e.g. above

+80°C), or by switching to an extreme pH, i.e. either alkaline (e.g. KOH or NaOH)

or acidic (e.g. perchloric acid, HCl or trichloroacetic acid)49. However, the most

widely used method is using cold methanol while maintaining the temperature below -20 °C, followed by collection of the cells with a centrifugation step at 0°C prior to lipid extraction44,48. Although quenching is performed to avoid the

(14)

2

enzymatic turnover of the analytes under study, the extreme physical-chemical conditions may lead to complications such as poor compatibility with the subsequent MS methods and more importantly, chemical degradation of some lipid species mainly through hydrolysis of the ester bonds28,50.

Quenching is a highly sample- and cell-dependent procedure. Differences in cellular composition (especially cell membrane and cell wall structure) and cell size, together with the ratio of methanol to water in the quenching solution may influence the efficiency of the procedure51. For lipidomics studies in cells,

quenching is a common procedure, whereas for biofluids it is generally omitted, since it is considered to induce changes in the samples28. However, in the case

of biofluids, sample alteration subsequent to collection is also possible due to residual enzymatic activity. Even in methanolic solutions, some enzymes are prone to remain active, thus, monitoring of substrates for enzymes (e.g., lutamyltransferase, alkaline phosphatase and urease) is recommended since it has provided useful insight into residual enzymatic activity during the preanalytical phase25,52.

Lipid extraction

Post collection sample handling and preparation are critical during the pre-analytical phase of lipidomics studies, since minor modifications in these processes may result in unwanted and unexpected changes, which can be confused with significant biochemical alterations53. For untargeted lipidomics

analyses, an ideal sample-preparation method should be (1) unselective, (2) simple and fast with a minimal number of steps and (3) reproducible. In contrast, an ideal sample-preparation method for targeted analysis should be (1) as selective as possible for the lipid(s) under study, (2) reproducible and (3) allow implementation of preconcentration and/or clean-up steps, if required28.

The necessity of disruption steps prior to lipid extraction makes the handling of solid (tissue) samples more complex in comparison to biofluids. The use of liquid nitrogen to snap-freeze the samples is a common procedure that minimizes stability issues and makes physical manipulation easier. The next step is homogenization of the sample, required to ensure efficient and reproducible extraction of the lipidome. Frozen solid samples can be homogenized with a mortar and pestle by manual grinding, or by using bead beater homogenizers together with an appropriate extraction solvent54. Sample handling of biofluids

is mainly focused on increasing the efficiency of lipid extraction by using different solvent systems, making it more straightforward than solid samples. As mentioned before, plasma/serum and urine are the most typical samples for

(15)

lipidomics studies. However, specialized fluids, such as tear fluid, cerebrospinal fluid and synovial fluid have recently been investigated as well (Table 1). Regardless of the type of sample, each matrix represents a different challenge for the analyst since its chemical composition (proteins, polar metabolites and lipid levels) determines the best way to proceed with the sample preparation. For instance, plasma and serum are lipid-rich biofluids, containing more than 17,000 species of lipids and fatty acids whose concentrations range between 4 pM to 2mM54–56. The high concentration of proteins is known to be the major

challenge for lipidomics in plasma and serum, as proteins are considered the main class of interfering compounds in these types of samples. Although the metabolome of human urine is rather complex, the lipidome is limited in complexity containing low concentrations (ranging from 0.1 to 1450 nM) of approximately 800 species of lipids and fatty acids57,58. Additionally, degradation

products of metabolites from food and/or drug intake represent a source of interference in urine samples. Therefore, whereas in plasma and serum, protein removal is important during sample preparation and is usually achieved by precipitation with the organic solvents used for lipid extraction, for urine, concentration processes such as lyophilization and/or solid-phase extraction must be considered due to the generally low concentration levels of lipids. The extraction of all lipid species in a comprehensive manner remains an active area of research in the lipidomics field. Liquid-liquid extraction is the standard technique for lipid analysis. The most commonly used methods for lipid extraction were introduced by Folch et al.59 and by Bligh and Dyer60. Over time,

these methods have been modified according to particular needs61,62, while

maintaining the basic principle of using different proportions of chloroform/ methanol that separate into an upper methanol-rich layer, containing hydrophilic compounds, and a lower chloroform-rich layer mainly containing lipids. Despite the popularity of these two methods, a major drawback is the need of retrieving the lipids from the lower chloroform-rich layer, which may lead to contamination that can compromise the analysis due to the presence of insoluble material

accumulating at the interphase2. To overcome this problem, two alternative

biphasic solvent systems were introduced more recently. Matyash et al.63

described the methyl tert-butyl ether (MTBE) extraction method and more recently Löfgren et al64 proposed the butanol-methanol (BUME) method. These

methods have the advantage that the lipid-rich organic phase forms the upper layer of the biphasic system. The extraction methods have been successfully applied for the extraction of key lipid classes, ranging from phospholipids and glycolipids to fatty acids, di- (DAGs) and tri-acylglycerols (TAGs). However,

(16)

2

contradictory reports particularly on the efficiency for extracting polar lipid species, such as glycolipids and lysophospholipids, have appeared4.

Since current lipidomics focuses on the use of high-throughput methods that allow the analysis of hundreds of samples in the shortest possible period of time (e.g. 96 samples each 3 hours according to Christinat et al. 2016)65,

single-phase lipid extractions that improve the recovery of polar lipid species while avoiding the inherent problems of the liquid-liquid extraction methods have recently gained in popularity4 (Table 1). By denaturing proteins that are later

removed by centrifugation, single-phase extractions focus on an “all-in-one-tube” approach eliminating the need for phase separation. Methanol, butanol, isopropanol and MTBE alone and miscible mixtures thereof have been used as solvents2,4,64. However, so far most studies using single-phase extraction

methods have focused on the targeted evaluation and recovery of a small set of lipid standards, hence, hindering the assessment of their suitability with respect to coverage of the lipidome in comparison to liquid-liquid extractions2,4.

Another important issue that has been rarely addressed regarding sample preparation procedures for lipidomics is the lack of standardized criteria to evaluate the suitability of different extraction methods. For targeted lipid analysis, this is typically achieved by validating the analytical method (from the sample processing to the data acquisition step), with respect to selectivity, accuracy, precision, recovery and linearity of the response for a small number of lipid species4,12. Given the complexity of the lipidome, it is not possible to

apply the same approach to evaluate the suitability of extraction methods for untargeted lipidomics analysis. However, some efforts have been made towards the standardization of a defined set of criteria. Previously, Sarafian et al.66 compared different single-phase and liquid-liquid extraction methods

with an untargeted approach. They concluded that simplicity of the extraction procedure, protein removal efficiency, repeatability, lipid coverage and lipid recovery were the most important requirements that an extraction method must fulfill in view of untargeted lipidomics. Based on these criteria, they found that using single-phase extractions mainly based on isopropanol (IPA) as solvent gave repeatable results of the lipidome from plasma samples with enhanced lipid coverage and good recovery. As precipitation of proteins with IPA is widely used in drug analysis, it may be argued that many rather polar compounds will also remain in the supernatant potentially complicating the analysis and affecting the final results. It is fair to say that, despite recent improvements, there is no extraction approach that provides a complete picture of the lipidome

(17)

Table 1. Ad vant ag es and dis ad vant ag es of e xt rac tion me thods c ommonl y used f or lipidomic s s tudie s (acc or din g t o T eo , e t al . 54). Ext rac tion me thod Type of s am ple A dv an tage s Dis ad vant ag es R ef er enc e Sin gle pha se ext rac tion •S er um (Biof luid ) •Sk in (S olid ) • E as y t o per for m • S hor t e xt rac tion t ime • L ow c os t • N o incr ea se in t em per at ur e f or t her molabile c om ponent s • N o e xt er nal ener gy supplied • Us ag e of t oxic or ganic sol vent • C an be har d t o r emo ve or ganic sol vent f or anal ysis 69,70 71 Liq uid-L iq uid ext rac tion •T ear s (Biof luid ) •S er um (Biof luid ) •P la sma (Biof luid ) •U rine (Biof luid ) • S yno vial fluid (Biof luid ) • A ther osc ler ot ic plaq ue (S olid ) •Sk in (S olid ) •T is sue (S olid ) • W ell-e st ablished me thod • E as y t o per for m • Che ap appar at us • S hor t e xt rac tion t ime • Che ap sol vent s used (e .g . c hlor of or m, me thanol , w at er) • N o incr ea sed t em per at ur e f or t her mall y labile c om pounds • N o e xt er nal ener gy supplied • Lar ge v olume of t oxic or ganic sol vent s • Usuall y needs mor e t han one type of sol vent • N eed t o r emo ve sol vent s t o av oid int er fer ence s w it h t he anal ysis 72 73–78 64,79–87 88,89 90 91 92–94 95,96 Solid pha se ext rac tion •S er um (Biof luid ) •Sput um (Biof luid ) •E ye (S olid ) •Sk in (S olid ) • E as y t o per for m • Cle ans up int er fer in g ma tr ix c om ponent s • Choice of dif fer ent SP E mode s • N o incr ea se in t em per at ur e f or t her mall y labile com pounds • N o e xt er nal ener gy supplied • S hor t e xt rac tion t ime • C os tl y SP E c ar tr id ge s • N eed t o w ash out or ganic sol vent s t o a void int er fer ence s w it h t he anal ysis • C an be har d t o r emo ve or ganic sol vent f or anal ysis 69,70,77 97 93 71,94

(18)

2

Super cr it ic al f luid ext rac tion •P la sma (Biof luid ) • N o or ganic sol vent • S hor t e xt rac tion t ime • Hi gh e xt rac tion e ff icienc y f or non-polar c om ponent s • CO 2 is r ec yc lable • T em per at ur e and pr es sur e c ont rollable • Modif ier s c an be added t o im pr ov e polar it y f or e xt rac tion ef ficienc y • Challen gin g t o oper at e • C os tl y • R eq uir es s pecif ic eq uipment 98 Micr ow av e-as sis ted ext rac tion •P la sma (Biof luid ) •Sk in (S olid ) • E as y t o per for m • S hor t e xt rac tion t ime • Hi gh e xt rac tion e ff icienc y • L ow c onsum pt ion of e xt rac tion sol vent • T em per at ur e and pr es sur e c ont rollable • P os sible los s of v ola tile or ganic com pounds • R eq uir es s pecif ic eq uipment • N eeds an e xt er nal sour ce of ener gy 99 100 Ult ra sound-as sis ted e xt rac tion •Blood (Biof luid ) • E as y t o per for m • S hor t e xt rac tion t ime • L ow e xt rac tion sol vent v olume used • T em per at ur e c ont rollable • H ar mf ul sol vent ma y be inhaled • N eeds an e xt er nal sour ce of ener gy • R eq uir es s pecif ic eq uipment • Incr ea sed t em per at ur e due t o fr ic tions c an po tent iall y alt er com pounds 5,101 Table 1. C ont inued

(19)

due to the structural diversity of its components, which makes it unlikely, that a ‘one size fits all’ approach will be developed any time soon if at all.

Solid phase extraction (SPE) is based on the use of a solid (stationary) phase and a liquid (mobile) phase to improve the selectivity of the analytical method, by capturing specific classes of lipids with similar physical-chemical properties54.

Consequently, this is a widespread methodology in targeted lipid analysis. SPE is a well-known sample-preparation method that typically requires a protein precipitation step and is combined with liquid-liquid extraction as additional clean-up step to enrich specific classes of lipids from the biological

matrix54. Normal-phase silica columns, reversed-phase columns (C8 and

C18), and ion-exchange columns (e.g. aminopropyl) are commonly used for the separation and sub-fractionation across a wide polarity range, from low polarity triacylglycerides (TAG) and cholesterol esters (CE) to more polar low molecular weight fatty acids and glycolipids67,68.

A number of alternative techniques that aid in the extraction process are being used more frequently. Supercritical fluid extraction (SFE), microwave-assisted extraction (MAE) and ultrasound-assisted extraction (UAE) are techniques that decrease extraction times and require lower amounts of solvents54. A summary

of the most common types of samples and extraction strategies used for lipidomics analysis is shown in Table 1.

Once lipid extracts are prepared, stability remains an issue that needs to be considered. At this point, in order to avoid degradation processes, dividing the extract into aliquots is recommended, as this will reduce the number of freeze-thaw cycles to which samples are subjected. Moreover, the removal of air from the headspace of sample vials containing the lipid extracts, by passing an inert gas, such as nitrogen, over the mouths of the vials prior to sealing them, can help to minimize lipid oxidation.

Common analytical platforms in lipidomics

Data acquisition is the next part of the lipidomics workflow once lipid extracts have been prepared (Figure 3). Below, we give a brief overview of the main analytical approaches that are currently used. More detailed accounts can be found in the literature1,54,102.

(20)

2

Robust analytical techniques that allow the simultaneous measurement of large numbers of lipids in a single biological sample form the basis of lipidomics.

Nuclear magnetic resonance spectroscopy (1H and 13C NMR) and mass

spectrometry (MS) are the most commonly applied techniques103.

NMR spectroscopy has been widely used since the early days of lipidomics research, notably for elucidating lipid structures102. The major advantages of

NMR-based lipid profiling are the high throughput combined with reproducibility and ease of analysis, besides high information content of the spectra due to simultaneous detection of all molecules in one snapshot104. In contrast, the

major drawback of this technique is its limited sensitivity.

Mass spectrometry (MS), combined with effective sample preparation and separation techniques, is undoubtedly the main approach in lipidomics due high sensitivity, specificity, as well as a wide dynamic range. Thus, hyphenated MS techniques have rapidly emerged as the dominant approach for lipid profiling105.

In conjunction with liquid chromatography (LC), ionization may be achieved using matrix assisted laser desorption/ionization (MALDI), atmospheric pressure chemical ionization (APCI) and electrospray ionization (ESI), the latter being the most popular, since it is a soft ionization technique that allows the generation of molecular ions without excessive fragmentation54. While

low-resolution mass analyzers like quadrupole ion traps or quadrupoles (IT, Q) are well-suited for targeted lipid analyses, they are suboptimal for lipidomics, due to the high complexity of lipid samples and the need for high mass resolution and mass accuracy. This is why time-of-flight (TOF) and orbital trap (Orbitrap) mass analyzers alone or in combination with IT or Q mass analyzers in hybrid or tandem configurations have become the instruments of choice for lipidomics, as they allow precursor ion selection and fragmentation to acquire MS/MS data of higher specificity106.

Despite the separation and identification capabilities of LC-high-resolution-MS for lipidomics analyses, there are important challenges that have to be addressed to get closer to a comprehensive view of a lipidome. Especially, the selection of the chromatographic conditions has an important effect on the type and number of lipids that can be detected. Most LC-MS-based lipidomics studies have been performed by reversed-phase high-performance liquid chromatography (RP-HPLC). Separation is governed by lipophilicity, which is determined by the length of the acyl chains. Thus, lipid species containing longer acyl chains elute later from the LC column than lipids with shorter acyl

(21)

chains, and lipids with saturated acyl chains elute later than their mono- or polyunsaturated analogs.

Hydrophilic interaction chromatography (HILIC) was more recently introduced for lipid analysis. This technique allows a separation of lipids primarily based on the hydrophilic nature of the head group. Utilization of HILIC in general has grown in popularity thanks to the excellent compatibility with mass spectrometry (enhanced MS sensitivity due to the high organic content in the mobile phase and the high efficiency of spray formation and desolvation)107. Since selectivity

of lipidome analyses by HILIC and RP is complementary, two-dimensional LC (LC x LC) methods have been established using a combination of both separation modes, thus increasing the coverage of the lipidome from the more polar lysophosphatidylcholines (lysoPC), lysophosphatidylethanolamines (lysoPE), phosphatidylcholines (PC), phosphatidylethanolamines (PE) and phosphatidylinositol (PI) species to the most apolar cholesterol esters (CE) and triglycerides (TG)54.

Conclusion

With this chapter, we focused on the pre-analytical aspects affecting lipidomics, we provide the reader with background information concerning lipidomics studies, and insights into some of the major limitations. Separation and detection techniques for lipidomics analysis are continuously evolving and being reviewed, but technical aspects related to the preanalytical stage related to stability issues of lipid species have rarely been addressed. Currently, lipidomics is moving towards its use as a diagnostic tool in a regulated clinical environment. Therefore, addressing lipid stability issues and avoiding the introduction of bias during the pre-analytical phase of the analysis, is critical for the further development of the field.

(22)

2

References

1. Sandra K, t’kindt R, Jorge L, Sandra P. The Art and Practice of Lipidomics. In: Metabolomics in Practice. Wiley-VCH Verlag GmbH & Co. KGaA; 2013:137-176. doi:10.1002/9783527655861.ch7

2. Löfgren L, Forsberg G-B, Ståhlman M, Tolley B, Tigyi GJ. The BUME method: a new rapid and simple chloroform-free method for total lipid extraction of animal tissue. Sci Rep. 2016;6(1):27688. doi:10.1038/srep27688

3. Garcia-Cazorla À, Mochel F, Lamari F, Saudubray J-M. The clinical spectrum of inherited diseases involved in the synthesis and remodeling of complex lipids. A tentative overview. J Inherit Metab

Dis. 2015;38(1):19-40. doi:10.1007/s10545-014-9776-6

4. Pellegrino RM, Di Veroli A, Valeri A, Goracci L, Cruciani G. LC/MS lipid profiling from human serum: A new method for global lipid extraction. Anal Bioanal Chem. 2014;406(30):7937-7948. doi:10.1007/ s00216-014-8255-0

5. Pizarro C, Arenzana-Rámila I, Pérez-Del-Notario N, Pérez-Matute P, González-Sáiz JM. Plasma lipidomic profiling method based on ultrasound extraction and liquid chromatography mass spectrometry. Anal Chem. 2013;85(24):12085-12092. doi:10.1021/ac403181c

6. Spener F, Lagarde M, Géloên A, Record M. Editorial: What is lipidomics? Eur J Lipid Sci Technol. 2003;105(9):481-482. doi:10.1002/ejlt.200390101

7. Han X, Gross RW. Global analyses of cellular lipidomes directly from crude extracts of biological samples by ESI mass spectrometry: a bridge to lipidomics. J Lipid Res. 2003;44(6):1071-1079. doi:10.1194/jlr.R300004-JLR200

8. McGhie TK, Rowan DD. Metabolomics for measuring phytochemicals, and assessing human and animal responses to phytochemicals, in food science. Mol Nutr Food Res. 2012;56:147-158. doi:10.1002/mnfr.201100545

9. Du F, Virtue A, Wang H, Yang X-F. Metabolomic analyses for atherosclerosis, diabetes, and obesity.

Biomark Res. 2013;1:1-17. doi:10.1186/2050-7771-1-17

10. Roberts LD, Souza AL, Gerszten RE, Clish CB. Targeted metabolomics. In: Ausubel FM, Brent R, Kingston RE, et al., eds. Current Protocols in Molecular Biology. Vol Chapter 30. John Wiley & Sons Inc.; 2012:30.2.1-24. doi:10.1002/0471142727.mb3002s98

11. Elkordy AA. Applications of Calorimetry in a Wide Context - Differential Scanning Calorimetry,

Isothermal Titration Calorimetry and Microcalorimetry. InTech; 2013. doi:10.5772/2898

12. U.S. Department of Health and Human Services, Food and Drug Administration (HSS-FDA). Guidance for Industry - Bioanalytical Method Validation.; 2013.

13. Rosenling T, Stoop MP, Smolinska A, et al. The impact of delayed storage on the measured proteome and metabolome of human cerebrospinal fluid. Clin Chem. 2011;57(12):1703-1711. doi:10.1373/ clinchem.2011.167601

14. Rosenling T, Slim CL, Christin C, et al. The effect of preanalytical factors on stability of the proteome and selected metabolites in Cerebrospinal Fluid (CSF). J Proteome Res. 2009;8(12):5511-5522. doi:10.1021/pr9005876

15. Heiskanen LA, Suoniemi M, Ta HX, Tarasov K, Ekroos K. Long-term performance and stability of molecular shotgun lipidomic analysis of human plasma samples. Anal Chem. 2013;85(18):8757-8763. doi:10.1021/ac401857a

16. Smilowitz JT, Zivkovic AM, Wan YJY, et al. Nutritional lipidomics: Molecular metabolism, analytics, and diagnostics. Mol Nutr Food Res. 2013;57(8):1319-1335. doi:10.1002/mnfr.201200808

17. Hyötyläinen T, Bondia-Pons I, Orešič M. Lipidomics in nutrition and food research. Mol Nutr Food Res. 2013;57(8):1306-1318. doi:10.1002/mnfr.201200759

18. Zhao Y-Y, Cheng X, Lin R-C. Lipidomics Applications for Discovering Biomarkers of Diseases in Clinical

Chemistry. Vol 313.; 2014. doi:10.1016/B978-0-12-800177-6.00001-3

19. Hyötyläinen T, Orešič M. Bioanalytical techniques in nontargeted clinical lipidomics. Bioanalysis. 2016;8(4):351-364. doi:10.4155/bio.15.244

(23)

20. Cajka T, Fiehn O. Comprehensive analysis of lipids in biological systems by liquid chromatography-mass spectrometry. TrAC - Trends Anal Chem. 2014;61:192-206. doi:10.1016/j.trac.2014.04.017 21. Satomi Y. Lipidomics for Pharmaceutical Research. In: Lipidomics: Technologies and Applications. ;

2012:319-328. doi:10.1002/9783527655946.ch16

22. Schörken U, Kempers P. Lipid biotechnology: Industrially relevant production processes. Eur J Lipid

Sci Technol. 2009;111(7):627-645. doi:10.1002/ejlt.200900057

23. Saude EJ, Sykes BD. Urine stability for metabolomic studies: effects of preparation and storage.

Metabolomics. 2007;3:19-27. doi:10.1007/s11306-006-0042-2

24. Dunn WB, Broadhurst D, Begley P, et al. Procedures for large-scale metabolic profiling of serum and plasma using gas chromatography and liquid chromatography coupled to mass spectrometry. Nat

Protoc. 2011;6:1060-1083. doi:10.1038/nprot.2011.335

25. Vuckovic D. Current trends and challenges in sample preparation for global metabolomics using liquid chromatography–mass spectrometry. Anal Bioanal Chem. 2012;403:1523-1548. doi:10.1007/ s00216-012-6039-y

26. Ishikawa M, Maekawa K, Saito K, et al. Plasma and serum lipidomics of healthy white adults shows characteristic profiles by subjects’ gender and age. PLoS One. 2014;9(3):1-12. doi:10.1371/journal. pone.0091806

27. Begum H, Li B, Shui G, et al. Discovering and validating between-subject variations in plasma lipids in healthy subjects. Sci Rep. 2016;6(1):19139. doi:10.1038/srep19139

28. Fernández-Peralbo MA, Luque de Castro MD. Preparation of urine samples prior to targeted or untargeted metabolomics mass-spectrometry analysis. TrAC - Trends Anal Chem. 2012;41:75-85. doi:10.1016/j.trac.2012.08.011

29. Byeon SK, Lee JY, Lee J-S, Moon MH. Lipidomic profiling of plasma and urine from patients with Gaucher disease during enzyme replacement therapy by nanoflow liquid chromatography– tandem mass spectrometry. J Chromatogr A. 2015;1381:132-139. doi:10.1016/j.chroma.2015.01.004 30. León Z, García-Cañaveras JC, Donato MT, Lahoz A. Mammalian cell metabolomics: experimental

design and sample preparation. Electrophoresis. 2013;34:2762-2775. doi:10.1002/elps.201200605 31. Rabinowitz JD. Cellular metabolomics of Escherchia coli. Expert Rev Proteomics. 2007;4:187-198.

doi:10.1586/14789450.4.2.187

32. Dunn WB, Winder CL. Sample preparation related to the intracellular metabolome of yeast: Methods for quenching, extraction, and metabolite quantitation. Methods Enzymol. 2011;500:277-297. doi:10.1016/B978-0-12-385118-5.00015-3

33. Dennis EA, Deems RA, Harkewicz R, et al. A mouse macrophage lipidome. J Biol Chem. 2010;285(51):39976-39985. doi:10.1074/jbc.M110.182915

34. Yin P, Peter A, Franken H, et al. Preanalytical aspects and sample quality assessment in metabolomics studies of human blood. Clin Chem. 2013;59:833-845. doi:10.1373/clinchem.2012.199257

35. Denery JR, Nunes AAK, Dickerson TJ. Characterization of differences between blood sample matrices in untargeted metabolomics. Anal Chem. 2011;83:1040-1047. doi:10.1021/ac102806p 36. O’Donnell VB, Murphy RC, Watson SP. Platelet lipidomics: Modern day perspective on lipid

discovery and characterization in platelets. Circ Res. 2014;114(7):1185-1203. doi:10.1161/ CIRCRESAHA.114.301597

37. Cella G, Girolami a, Sasahara a a. Platelet activation with unfractionated heparin at therapeutic concentrations and comparison with low-molecular-weight heparin and with a direct thrombin inhibitor. Circulation. 1999;99(25):3323. doi:10.1161/01.CIR.99.25.3323

38. Gonzalez-Covarrubias V, Dane A, Hankemeier T, Vreeken RJ. The influence of citrate, EDTA, and heparin anticoagulants to human plasma LC-MS lipidomic profiling. Metabolomics. 2013;9(2):337-348. doi:10.1007/s11306-012-0450-4

39. Wedge DC, Allwood JW, Dunn W, et al. Is serum or plasma more appropriate for intersubject comparisons in metabolomic studies? An assessment in patients with small-cell lung cancer. Anal

(24)

2

40. Mei H, Hsieh Y, Nardo C, et al. Investigation of matrix effects in bioanalytical high-performance

liquid chromatography/tandem mass spectrometric assays: application to drug discovery. Rapid

Commun Mass Spectrom. 2003;17(1):97-103. doi:10.1002/rcm.876

41. Dimeski G, Mollee P, Carter A. Increased lipid concentration is associated with increased hemolysis [5]. Clin Chem. 2005;51(12):2425. doi:10.1373/clinchem.2005.058644

42. Silvester S, Zang F. Overcoming non-specific adsorption issues for AZD9164 in human urine samples: Consideration of bioanalytical and metabolite identification procedures. J Chromatogr B. 2012;893-894:134-143. doi:10.1016/j.jchromb.2012.03.004

43. Emwas A-H, Roy R, McKay RT, et al. Recommendations and Standardisation of Biomarker Quantification Using NMR Based Metabolomics with Particular Focus on Urinary Analysis. J

Proteome Res. 2016;15:360-373.

44. Dettmer K, Nürnberger N, Kaspar H, Gruber MA, Almstetter MF, Oefner PJ. Metabolite extraction from adherently growing mammalian cells for metabolomics studies: optimization of harvesting and extraction protocols. Anal Bioanal Chem. 2011;399:1127-1139. doi:10.1007/s00216-010-4425-x 45. Duarte IF, Marques J, Ladeirinha AF, et al. Analytical approaches toward successful human cell

metabolome studies by NMR spectroscopy. Anal Chem. 2009;81:5023-5032. doi:10.1021/ac900545q 46. Villas-Bôas SG, Bruheim P. Cold glycerol-saline: The promising quenching solution for accurate

intracellular metabolite analysis of microbial cells. Anal Biochem. 2007;370:87-97. doi:10.1016/j. ab.2007.06.028

47. Villas-Bôas SG, Højer-Pedersen J, Akesson M, Smedsgaard J, Nielsen J. Global metabolite analysis of yeast: evaluation of sample preparation methods. Yeast. 2005;22:1155-1169. doi:10.1002/yea.1308 48. Bolten CJ, Kiefer P, Letisse F, Portais J-C, Wittmann C. Sampling for metabolome analysis of

microorganisms. Anal Chem. 2007;79:3843-3849. doi:10.1021/ac0623888

49. Mashego MR, Rumbold K, De Mey M, Vandamme E, Soetaert W, Heijnen JJ. Microbial metabolomics: Past, present and future methodologies. Biotechnol Lett. 2007;29(1):1-16. doi:10.1007/s10529-006-9218-0

50. Aveldaño MI, Horrocks LA. Quantitative release of fatty acids from lipids by a simple hydrolysis procedure. J Lipid Res. 1983;24(8):1101-1105.

51. Sellick CA, Knight D, Croxford AS, et al. Evaluation of extraction processes for intracellular metabolite profiling of mammalian cells: matching extraction approaches to cell type and metabolite targets.

Metabolomics. 2010;6:427-438. doi:10.1007/s11306-010-0216-9

52. Bernini P, Bertini I, Luchinat C, Nincheri P, Staderini S, Turano P. Standard operating procedures for pre-analytical handling of blood and urine for metabolomic studies and biobanks. J Biomol NMR. 2011;49(3-4):231-243. doi:10.1007/s10858-011-9489-1

53. Theodoridis GA, Gika HG, Want EJ, Wilson ID. Liquid chromatography-mass spectrometry based global metabolite profiling: A review. Anal Chim Acta. 2012;711:7-16. doi:10.1016/j.aca.2011.09.042 54. Teo CC, Chong WPK, Tan E, Basri NB, Low ZJ, Ho YS. Advances in sample preparation and analytical

techniques for lipidomics study of clinical samples. TrAC - Trends Anal Chem. 2015;66:1-18. doi:10.1016/j.trac.2014.10.010

55. Psychogios N, Hau DD, Peng J, et al. The human serum metabolome. PLoS One. 2011;6(2). doi:10.1371/journal.pone.0016957

56. Trabado SV, Al-Salameh A, Croixmarie V, et al. The human plasma-metabolome: Reference values in 800 French healthy volunteers; impact of cholesterol, gender and age. doi:10.1371/journal. pone.0173615

57. Bouatra S, Aziat F, Mandal R, et al. The Human Urine Metabolome. PLoS One. 2013;8(9). doi:10.1371/ journal.pone.0073076

58. Rockwell HE, Gao F, Chen EY, et al. Dynamic Assessment of Functional Lipidomic Analysis in Human Urine. Lipids. 2016;51(7):875-886. doi:10.1007/s11745-016-4142-0

59. Folch J, Lees M, Sloane-Stanley G. A simple method for the isolation and purification of total lipids from animal tissues. J biol Chem. 1957;226(3):497-509. doi:10.1007/s10858-011-9570-9

(25)

60. BLIGH EG, DYER WJ. A rapid method of total lipid extraction and purification. Can J Biochem Physiol. 1959;37(8):911-917. doi:10.1139/o59-099

61. Retra K, Bleijerveld O, Van Gestel R, Tielens AG, Van Hellemond J, Brouwers J. A simple and universal method for the separation and identification of phospholipid molecular species. Rapid Commun

Mass Spectrom. 2008;22(12):1853-1862. doi:10.1002/rcm.3562

62. Reis a., Rudnitskaya a., Blackburn GJ, Fauzi NM, Pitt a. R, Spickett CM. A comparison of five lipid extraction solvent systems for lipidomic studies of human LDL. J Lipid Res. 2013;54(7):1812-1824. doi:10.1194/jlr.M034330

63. Matyash V, Liebisch G, Kurzchalia T V, Shevchenko A, Schwudke D. Lipid extraction by methyl-tert-butyl ether for high-throughput lipidomics. J Lipid Res. 2008;49(5):1137-1146. doi:10.1194/jlr. D700041-JLR200

64. Löfgren L, Ståhlman M, Forsberg G-BG-B, et al. The BUME method: a novel automated chloroform-free 96-well total lipid extraction method for blood plasma. J Lipid Res. 2012;53(8):1690-1700. doi:10.1194/jlr.D023036

65. Christinat N, Morin-Rivron D, Masoodi M. High-Throughput Quantitative Lipidomics Analysis of Nonesterified Fatty Acids in Human Plasma. J Proteome Res. 2016;15(7):2228-2235. doi:10.1021/acs. jproteome.6b00198

66. Sarafian MH, Gaudin M, Lewis MR, et al. Objective set of criteria for optimization of sample preparation procedures for ultra-high throughput untargeted blood plasma lipid profiling by ultra performance liquid chromatography-mass spectrometry. Anal Chem. 2014;86(12):5766-5774. doi:10.1021/ac500317c

67. Hamilton JG, Comai K. Rapid separation of neutral lipids, free fatty acids and polar lipids using prepacked silica Sep-Pak columns. Lipids. 1988;23(12):1146-1149. doi:10.1007/BF02535281 68. Kim H, Salem N. Separation of lipid classes by solid phase extraction. J Lipid Res.

1990;31(3):2285-2289.

69. Stübiger G, Aldover-Macasaet E, Bicker W, et al. Targeted profiling of atherogenic phospholipids in human plasma and lipoproteins of hyperlipidemic patients using MALDI-QIT-TOF-MS/MS.

Atherosclerosis. 2012;224(1):177-186. doi:10.1016/j.atherosclerosis.2012.06.010

70. Zhao Z, Xu Y. An extremely simple method for extraction of lysophospholipids and phospholipids from blood samples. J Lipid Res. 2010;51(3):652-659. doi:10.1194/jlr.D001503

71. T’Kindt R, Jorge L, Dumont E, et al. Profiling and characterizing skin ceramides using reversed-phase liquid chromatography-quadrupole time-of-flight mass spectrometry. Anal Chem. 2012;84(1):403-411. doi:10.1021/ac202646v

72. Lam SM, Tong L, Duan X, Petznick A, Wenk MR, Shui G. Extensive characterization of human tear fluid collected using different techniques unravels the presence of novel lipid amphiphiles. J Lipid

Res. 2014;55(2):289-298. doi:10.1194/jlr.M044826

73. Cui L, Lee YH, Kumar Y, et al. Serum Metabolome and Lipidome Changes in Adult Patients with Primary Dengue Infection. PLoS Negl Trop Dis. 2013;7(8). doi:10.1371/journal.pntd.0002373 74. Heilbronn LK, Coster ACF, Campbell L V., et al. The effect of short-term overfeeding on serum lipids

in healthy humans. Obesity. 2013;21(12). doi:10.1002/oby.20508

75. Hilvo M, Gade S, Hyotylainen T, et al. Monounsaturated fatty acids in serum triacylglycerols are associated with response to neoadjuvant chemotherapy in breast cancer patients. Int J Cancer. 2014;134(7):1725-1733. doi:10.1002/ijc.28491

76. Montoliu I, Scherer M, Beguelin F, et al. Serum profiling of healthy aging identifies phospho- and sphingolipid species as markers of human longevity. Aging (Albany NY). 2014;6(1):9-25. doi:10.18632/aging.100630

77. Chen Y, Liu Q, Yong S, Teo HL, Lee TK. An improved reference measurement procedure for triglycerides and total glycerides in human serum by isotope dilution gas chromatography-mass spectrometry. Clin Chim Acta. 2014;428:20-25. doi:10.1016/j.cca.2013.10.014

(26)

2

78. Zivkovic AM, Wiest MM, Nguyen UT, Davis R, Watkins SM, German JB. Effects of sample handling

and storage on quantitative lipid analysis in human serum. Metabolomics. 2009;5(4):507-516. doi:10.1007/s11306-009-0174-2

79. Sandra K, Pereira A dos S, Vanhoenacker G, David F, Sandra P. Comprehensive blood plasma lipidomics by liquid chromatography/quadrupole time-of-flight mass spectrometry. J Chromatogr

A. 2010;1217(25):4087-4099. doi:10.1016/j.chroma.2010.02.039

80. Chen F, Maridakis V, O’Neill E a, et al. The effects of simvastatin treatment on plasma lipid-related biomarkers in men with dyslipidaemia. Biomarkers. 2011;16(December 2010):321-333. doi:10.3109 /1354750X.2011.561367

81. Ollero M, Astarita G, Guerrera IC, et al. Plasma lipidomics reveals potential prognostic signatures within a cohort of cystic fibrosis patients. J Lipid Res. 2011;52(5):1011-1022. doi:10.1194/jlr.P013722 82. Shah V, Castro-Perez JM, McLaren DG, Herath KB, Previs SF, Roddy TP. Enhanced data-independent

analysis of lipids using ion mobility-TOFMSE to unravel quantitative and qualitative information in human plasma. Rapid Commun Mass Spectrom. 2013;27(19):2195-2200. doi:10.1002/rcm.6675 83. Lankinen M, Schwab U, Erkkilä A, et al. Fatty fish intake decreases lipids related to inflammation and

insulin signaling - A lipidomics approach. PLoS One. 2009;4(4). doi:10.1371/journal.pone.0005258 84. Graessler J, Schwudke D, Schwarz PEH, Herzog R, Schevchenko A, Bornstein SR. Top-down

lipidomics reveals ether lipid deficiency in blood plasma of hypertensive patients. PLoS One. 2009;4(7). doi:10.1371/journal.pone.0006261

85. Gürdeniz G, Rago D, Bendsen NT, Savorani F, Astrup A, Dragsted LO. Effect of trans Fatty Acid Intake on LC-MS and NMR Plasma Profiles. PLoS One. 2013;8(7). doi:10.1371/journal.pone.0069589 86. Zhou X, Mao J, Ai J, et al. Identification of Plasma Lipid Biomarkers for Prostate Cancer by Lipidomics

and Bioinformatics. PLoS One. 2012;7(11). doi:10.1371/journal.pone.0048889

87. Bui HH, Leohr JK, Kuo M-S. Analysis of sphingolipids in extracted human plasma using liquid chromatography electrospray ionization tandem mass spectrometry. Anal Biochem. 2012;423(2):187-194. doi:10.1016/j.ab.2012.01.027

88. Min HK, Lim S, Chung BC, Moon MH. Shotgun lipidomics for candidate biomarkers of urinary phospholipids in prostate cancer. Anal Bioanal Chem. 2011;399(2):823-830. doi:10.1007/s00216-010-4290-7

89. Kim H, Ahn E, Moon MH. Profiling of human urinary phospholipids by nanoflow liquid chromatography/tandem mass spectrometry. Analyst. 2008;133(12):1656. doi:10.1039/b804715d 90. Giera M, Ioan-Facsinay A, Toes R, et al. Lipid and lipid mediator profiling of human synovial fluid

in rheumatoid arthritis patients by means of LC–MS/MS. Biochim Biophys Acta - Mol Cell Biol Lipids. 2012;1821(11):1415-1424. doi:10.1016/j.bbalip.2012.07.011

91. Stegemann C, Drozdov I, Shalhoub J, et al. Comparative lipidomics profiling of human atherosclerotic plaques. Circ Cardiovasc Genet. 2011;4(3):232-242. doi:10.1161/CIRCGENETICS.110.959098 92. van Smeden J, Hoppel L, van der Heijden R, Hankemeier T, Vreeken RJ, Bouwstra JA. LC/MS analysis

of stratum corneum lipids: ceramide profiling and discovery. J Lipid Res. 2011;52(6):1211-1221. doi:10.1194/jlr.M014456

93. Acar N, Berdeaux O, Grégoire S, et al. Lipid composition of the human eye: Are red blood cells a good mirror of retinal and optic nerve fatty acids? PLoS One. 2012;7(4). doi:10.1371/journal.pone.0035102 94. Shin JH, Shon JC, Lee K, et al. A lipidomic platform establishment for structural identification of skin

ceramides with non-hydroxyacyl chains. Anal Bioanal Chem. 2014;406(7):1917-1932. doi:10.1007/ s00216-013-7601-y

95. Cheng H, Wang M, Li JL, Cairns NJ, Han X. Specific changes of sulfatide levels in individuals with pre-clinical Alzheimer’s disease: An early event in disease pathogenesis. J Neurochem. 2013;127(6):733-738. doi:10.1111/jnc.12368

96. Pietiläinen KH, Róg T, Seppänen-Laakso T, et al. Association of Lipidome Remodeling in the Adipocyte Membrane with Acquired Obesity in Humans. Tieleman DP, ed. PLoS Biol. 2011;9(6):e1000623. doi:10.1371/journal.pbio.1000623

(27)

97. Cha D, Cheng D, Liu M, Zeng Z, Hu X, Guan W. Analysis of fatty acids in sputum from patients with pulmonary tuberculosis using gas chromatography-mass spectrometry preceded by solid-phase microextraction and post-derivatization on the fiber. J Chromatogr A. 2009;1216(9):1450-1457. doi:10.1016/j.chroma.2008.12.039

98. Uchikata T, Matsubara A, Fukusaki E, Bamba T. High-throughput phospholipid profiling system based on supercritical fluid extraction–supercritical fluid chromatography/mass spectrometry for dried plasma spot analysis. J Chromatogr A. 2012;1250:69-75. doi:10.1016/j.chroma.2012.06.031 99. Morais DR de, Visentainer JEL, Santos LP dos, Matsushita M, Souza NE de, Visentainer JV. Evaluation

of lipid extraction and fatty acid composition of human plasma. Rev Bras Hematol Hemoter. 2010;32(6):439-443. doi:10.1590/S1516-84842010000600006

100. González-Illán F, Ojeda-Torres G, Díaz-Vázquez LM, Rosario O. Detection of fatty acid ethyl esters in skin surface lipids as biomarkers of ethanol consumption in alcoholics, social drinkers, light drinkers, and teetotalers using a methodology based on microwave-assisted extraction followed by solid-phase microextrac. J Anal Toxicol. 2011;35(4):232-237.

101. Pang LQ, Liang QL, Wang YM, Ping L, Luo GA. Simultaneous determination and quantification of seven major phospholipid classes in human blood using normal-phase liquid chromatography coupled with electrospray mass spectrometry and the application in diabetes nephropathy. J Chromatogr B

Anal Technol Biomed Life Sci. 2008;869(1-2):118-125. doi:10.1016/j.jchromb.2008.05.027

102. Gross RW, Han X. Lipidomics at the interface of structure and function in systems biology. Chem

Biol. 2011;18(3):284-291. doi:10.1016/j.chembiol.2011.01.014

103. Madsen R, Lundstedt T, Trygg J. Chemometrics in metabolomics-A review in human disease diagnosis. Anal Chim Acta. 2010;659:23-33. doi:10.1016/j.aca.2009.11.042

104. Spagou K, Tsoukali H, Raikos N, Gika H, Wilson ID, Theodoridis G. Hydrophilic interaction chromatography coupled to MS for metabonomic/metabolomic studies. J Sep Sci. 2010;33:716-727. doi:10.1002/jssc.200900803

105. Bennette NB, Eng JF, Dismukes GC. An LC-MS-based chemical and analytical method for targeted metabolite quantification in the model cyanobacterium Synechococcus sp. PCC 7002. Anal Chem. 2011;83:3808-3816. doi:10.1021/ac200108a

106. Lu W, Bennett BD, Rabinowitz JD. Analytical strategies for LC-MS-based targeted metabolomics. J

Chromatogr B Anal Technol Biomed Life Sci. 2008;871:236-242. doi:10.1016/j.jchromb.2008.04.031

107. Chen J, Wang W, Lv S, et al. Metabonomics study of liver cancer based on ultra performance liquid chromatography coupled to mass spectrometry with HILIC and RPLC separations. Anal Chim Acta. 2009;650:3-9. doi:10.1016/j.aca.2009.03.039

(28)
(29)

Referenties

GERELATEERDE DOCUMENTEN

Lipids are taking essential roles in energy storage, signal transduction, membrane construction and more, therefore, lipidomics strategies are essential to gain insight in

model representative for CN-1, showed that transplantation of the small intestine from Wistar rats to Gunn rats decreased serum bilirubin levels in the latter, demonstrating that

Here we assessed the bilirubin and lipid phenotype in wild type, heterozygous and homozygous Gunn-Ugt1a1 j/BluHsdRrrc rat littermates in neonatal and adult conditions

increase in fecal UCB and subsequent decrease in plasma UCB levels upon higher intestinal fat concentrations is the result of UCB ‘capturing’ by fatty acids, meaning that

These effects were characterized by increased plasma levels of FGF21, browning of subcutaneous white adipose tissue, increased body temperature and energy expenditure, while

In ApoE-deficient mice, administration of the phenolic acid ferulic acid (FA) increased the hepatic levels of AhR which was associated with a decreased hepatic expression

Bovendien had activering van LXR en FXR geen hypobilirubinemische effecten in Gunn ratten, terwijl LXR activatie zelfs de plasma totale bilirubine (TB) waarden verhoogde,

Disorders of bilirubin and lipid metabolism: models and targets of intervention.. University