Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Nov 1.
Published in final edited form as: Biochim Biophys Acta. 2011 Jun 13;1811(11):724–736. doi: 10.1016/j.bbalip.2011.06.005

High-Throughput Lipidomic Analysis of Fatty Acid Derived Eicosanoids and N-Acylethanolamines

Darren S Dumlao 1,*, Matthew W Buczynski 1,*, Paul C Norris 1, Richard Harkewicz 1, Edward A Dennis 1
PMCID: PMC3205334  NIHMSID: NIHMS312202  PMID: 21689782

1. Introduction

Eicosanoids and N-acylethanolamines (NAEs) are very important bioactive lipid molecules that signal numerous physiological processes [1, 2]. Comprehensive metabolomic tools to study these lipids and their biological involvement have been challenging since lipids represent a very diverse group of molecules comprised of many different classes and subclasses [3]. Additionally, each subclass has many distinct chemical features making it very difficult to monitor every type of lipid in a single analysis. LIPID Metabolites And Pathways Strategy (LIPID MAPS), a NIH-funded consortium was created to develop the infrastructure required including the specific methodology reported herein, an extensive lipid database, and to develop lipid standards (www.lipidmaps.org).

Eicosanoids and NAEs comprise two classes of important bioactive lipid signaling molecules that act through binding to their cognate receptors. Eicosanoids and NAEs play a key role in the innate immune system modulating inflammation, cellular recruitment, pain signaling, blood pressure response, and fever [1, 2]. Additionally, many of these lipid metabolites have been implicated in a wide range of complex disease pathologies including cancer [4, 5], atherosclerosis [6], rheumatoid arthritis [7], cystic fibrosis [8] and neurodegeneration [9].

Eicosanoids represent a large diverse group of lipids, in part, due to nonspecific synthases that can utilize different polyunsaturated fatty acids (PUFA) as substrates. The complexity of the eicosanoids is further complicated because these molecules can then act as substrates for other synthases, either through an intracellular or trans-cellular mechanism [2, 10]. They are derived from polyunsaturated fatty acids (typically arachidonic acid) located at the sn-2 position of membrane glycerophospholipids liberated by enzymes with phospholipase A2 (PLA2) activity [11, 12]. Group IVA cPLA2 has been thought to be the main phospholipase responsible for the fatty acyls liberated from membrane phospholipids, while a recent report suggest that MAGL is the main lipase responsible for this activity in brain [13, 14]. These free fatty acyls serve as substrates for cyclooxygenases (COX), lipoxygenases (LOX), and cytochrome P450s (CYP) enzymes [1, 2, 15, 16]. Most eicosanoid studies have just focused on prostaglandin E2 (PGE2) and the role it plays in inflammatory responses. Since many eicosanoids display redundant signaling properties, efforts have been made to study these mediators collectively.

NAEs represent a class of endogenous bioactive signaling lipids composed of a fatty acyl conjugated to ethanolamine through the amide bond [17, 18]. The arachidonoyl species, anandamide (AEA), has received the most attention due to its anti-inflammatory action, nearly all endogenous fatty acyl species have been detected as ethanolamides in vivo [19]. A number of different pathways have been implicated in NAE formation, but the specific enzymes generating synaptic AEA formation remains unclear. To date, three main enzymatic routes for the formation of AEA from n-acyl phosphatidylethanolamines (NAPE) have been identified: (1) hydrolysis by a NAPE-specific phospholipase D (NAPE-PLD), (2) sequential phospholipase A/B activity of ABHD4, followed by a metal-dependent phosphodiesterase, and (3) sequential phospholipase C (PLC) and phosphatase activity. Inactivation of NAE signaling occurs primarily through their hydrolysis to form the free fatty acids and ethanolamine. Fatty acid acyl hydrolase (FAAH) has been identified as the primary means of AEA metabolism through a well-characterized serine hydrolase mechanism. Predominately, FAAH associates with intracellular membranes such as the endoplasmic reticulum and the Golgi apparatus. A great number of small molecules have been developed as selective inhibitors of FAAH [20-23], which has been a hotly pursued pharmacological target.

The quantitation of eicosanoids has been a challenging task due to the number of chemically different yet structurally similar metabolites (over 100). In the past, enzyme-linked immunosorbent assays (EIA) were employed to monitor a single eicosanoid species [24, 25]. Due to the lack of commercially available antibodies and their non-specificity, this approach was severely limited. Additionally, the EIA approach lacked the robustness to perform large scale analyses. The technological advancements in mass spectrometry and its application to monitor eicosanoids have led to a robust foundation for eicosanoid research. Using gas chromatography mass spectrometry (GC/MS) allowed for many eicosanoids to be monitored simultaneously, however, a lone chemical derivatization agent was not suitable for all eicosanoids to be monitored in a single analysis [26]. Also, GC/MS was not suited for monitoring every type of eicosanoid species [27]. Electrospray-ionization tandem mass spectrometry (ESI-MS/MS), which does not require a prior derivatization step, has become a staple in eicosanoid biology since it was first employed by Margalit and colleagues for simultaneously monitoring 14 different eicosanoid species in a single analysis [28]. The number of distinct eicosanoids that can be monitored in a single analysis has been steadily increasing as more pure standards have become commercially available, as well as improvements in mass spectrometer hardware and data analysis software.

Previously, we reported on the use of GC/MS to analyze free fatty acids [29] and LC/MS/MS to analyze eicosanoids [30]. Our initial eicosanoid methodology was capable of monitoring 60 unique species in a single 16 min analysis. Since our initial report, significant improvements have been made which more than doubles the number of eicosanoids monitored (141) and quantified (100). Additionally, we have applied this technique toward a separate methodology capable of monitoring 36 NAE metabolites. Here, we present the design and rationale behind our high-throughput LC/MS/MS methodologies for monitoring and quantitating eicosanoid and NAE metabolites, and highlight the improvements made over our previous technique. Also, we provide an example of the application of our methodology.

2. Materials and Methods

2.1. Sample Preparation

The same sample preparation is used when analyzing eicosanoids or NAEs. All samples were resuspended in 1.0 ml of 10% methanol water (v/v). Tissue samples were subjected to sonication for 6 seconds to break up any connective tissue. Samples were spiked with 50 μL of a 50 pg/ μL (2.5 ng total) deuterated internal standard solution. Lipid metabolites were extracted using strata-x 33 u polymerized solid reverse phase extraction columns (Phenomenex, CA; cat # 8B-S100-UBJ) as indicated by manufacturer's directions. Briefly, columns were washed with 3.5 ml of 100% methanol, followed by 3.5 ml of water before samples were extracted. Samples were washed with 3.5 ml of 10% methanol to remove non-specific binding metabolites. Lipids were eluted into 1.0 ml of methanol and stored at -80C° before being analyzed to prevent metabolite degradation.

2.2. High perfromance liquid chromatography (HPLC)

Both eicosanoid and NAE samples are subjected to the same treatment for HPLC analysis, although different buffer systems are employed. Extracted samples in 100% methanol are lyophilized to dryness using a speed-vac concentrator (Savant, model # SC110-120), and resuspended in 90 μL of their respective solvent A. For the eicosanoids methodology, solvent AEICOS consists of water-acetonitrile-acetic acid (70:30:0.02; v/v/v), while solvent BEICOS consists of acetonitrile-isopropyl alcohol (50:50, v/v). For the NAE methodology, solvent ANAE consists of water-acetonitrile-acetic acid ((70:30:0.1; v/v/v) + 1 g/L ammonium acetate) and solvent BNAE consists of acetonitrile-isopropyl alcohol-acetic acid ((45:45:10; v/v/v) + 1 g/L ammonium acetate). Samples can be subjected to each methodology alone or analyzed together in series. After a sample has been analyzed for eicosanoids, the presence of NAEs can be determined with the addition of 10 μL of a water-acetonitrile-acetic acid (70:30:0.1; v/v/v) + 5 g/L ammonium acetate solution that makes the sample suitable to be analyzed in positive-ion mode.

An aliquot of 40 μL of sample injected on the HPLC system was the standard amount routinely analyzed. Eicosanoids were separated on a Synergi reverse-phase C18 column (2.1 × 250 mm; Phenomenex, CA) and NAEs were separated on a Luna reverse-phase C8 column (2.1 mm · 250 mm, Phenomenex, CA). Both sample types use a flow rate of 300 μL/min at 50°C. The gradient program used to separate the eicosanoids is as follows: 1 min (0% solvent BEICOS), 3 min (25% solvent BEICOS), 11 min (45% solvent BEICOS), 13 min (60% solvent BEICOS), 18 min (75% solvent BEICOS), 18.5 min (90% solvent BEICOS), 20 min (90% solvent BEICOS), 21 min (0% solvent BEICOS). A linear gradient was maintained between each step. The column was re-equilibrated by holding 0% solvent BEICOS between min 21 to 25 before the next sample injection. The gradient program used to separate the NAEs is as follows: 1 min (0% solvent BNAE), 4 min (50% solvent BNAE), 14 min (100% solvent BNAE), 20 min (100% solvent BNAE), and 21 min (0% solvent BNAE). A linear gradient was maintained between each step. The column was re-equilibrated by holding 0% solvent BNAE between min 21 to 25 before the next sample injection.

2.3. Mass Spectrometry

An ABI/Sciex (Foster City, CA) 4000 QTRAP hybrid, triple quadrupole, linear ion trap mass spectrometer equipped with a Turbo V ion source was used for all mass spectrometry analysis. Analyst 1.5 software package was used to operate the mass spectrometer. Nitrogen gas was used as the collision gas for all metabolites. Eicosanoids were detected in negative electrospray ion mode with the following source parameters: CUR = 10 psi, GS1 = 30psi, GS2 = 30 psi, IS = -4500 V, CAD = HIGH, TEMP = 525°, ihe = ON, EP = -10 V, and CXP = -10 V. NAEs were analyzed in positive electrospray ion mode with the following source parameters: CUR = 10 psi, GS1 = 30psi, GS2 = 30 psi, IS = -4500 V, CAD = HIGH, TEMP = 525°, ihe = ON, EP = -10 V, and CXP = -10 V.

2.4. Quantitation

Eicosanoids and NAEs are quantitated using a stable isotope dilution technique. Primary standard curves are produced separately for eicosanoids and NAEs. Primary standard curves are generated from mixed primary standard stocks at 7 different concentrations (0.1 ng, 0.3 ng, 1 ng, 3 ng, 10 ng, 30 ng, and 100 ng). An aliquot of 50 mL of primary standard stock with the concentration ranging from 1 pg/μL (0.1 ng stock) to 1000 pg/μL (100 ng) are added to an aliquot of 25 μL of internal standard stock. For eicosanoids, 75 μL of 0.2% acetic acid-water were added, while for NAEs 75 μL of solvent ANAE (water-acetonitrile-acetic acid (70:30:0.1; v/v/v) + 1 g/L ammonium acetate). A 40 μL aliquot of each primary standard mix is analyzed with mass spectrometric methodologies. Primary standards were run in duplicate and averaged. Primary standard curves are determined by generating a linear regression trend line that is forced through 0. The mulitquant 1.1 software package (ABI-Sciex) was used to quantitate all metabolites.

3. Rationale and Discussion

3.1. Sample preparation

These methodologies have been applied to media from cultured primary cells, cultured cell lines (RAW264.7 and HEK293) [31], rat spinal cord tissue [19], murine papillomas, and murine tibiotarsal ankle joints [32]. The same sample preparation is used when analyzing eicosanoids or NAEs. All tissue samples were disrupted with a probe sonicator, while murine papilloma tissue required homogenization prior to sonication. Lipid metabolites were isolated from cultured media and tissue samples using solid phase extraction, except for murine ankle joints where a prior liquid / liquid extraction was employed [32]. A murine ankle joint sample is comprised mostly of bone and required an additional total lipid extraction.

Purification using flash chromatography can significantly enhance lipid detection and quantization limits by removing other chemical species that can diminish the overall sensitivity though processes such as ion suppression. Different extraction methods have been employed in the study of these lipid mediators [33, 34]. We decided to use a solid phase extraction technique, which is more suitable for processing a large number of samples than a more efficient liquid/liquid extraction technique as described in Golovko et al., 2008 [33]. The solid phase extraction technique utilizes Strata-x polymerized solid reverse phase extraction columns (Phenomenex, CA; cat # 8B-S100-UBJ). Samples are applied to these columns in 10% methanol, a concentration of organic solvent empirically determined to be sufficient to solubilize the eicosanoids and NAEs in our assay, yet not high enough to prevent them from binding to the column. An additional column volume of 10% methanol is used as a wash, which serves to elute non-specfic hydrophobic chemical species and salts from the sample. Subsequently, eicosanoids and NAEs are eluted off the column with 100% of methanol; however, a large fraction of the more lipophilic species that could potentially lead to ion suppression is retained. Previously, our lab has reported on some lipid recoveries obtained from this extraction technique [30]. Lipids are stored in 100% methanol at -80C° to minimize non-enzymatic oxidation and degradation until analysis.

A crucial aspect of our methodology is the usage of deuterated internal standards. All samples are spiked with a deuterated internal standard solution prior to lipid extraction. The use of deuterated internal standards is intrinsic to our ability to quantitate these lipids, and is covered in greater depth in the quantitation section (3.5). Deuterated internal standards also serve an important role in the extraction and storage process. Since a deuterated internal standard is either an analogous lipid metabolite or a molecule with similar chemical characteristics, both lipid metabolite and internal standard will have similar extraction efficiencies and rates of degradation. Thus, any amount of a lipid metabolite that is lost due to the extraction process or degradation will be accounted for.

3.2. Method design

Our methodology uses a targeted approach to identify and quantitate lipids using mass spectrometry (MS) coupled with high performance liquid chromatography (HPLC). This approach is known as the Comprehensive Lipidomic Analysis by Separation Simplification (CLASS) [3], where lipid metabolites are separated based on their chemical properties prior to mass analysis, then monitored by collision-induced decomposition (CID) in conjunction with electrospray ionization tandem mass spectrometry (ESI-MS/MS) [3]. Using CID, whereby each lipid metabolite creates ion fragments unique to its structural components, they can be subsequently measured by monitoring one of these ion fragments. A targeted MS approach allows for a higher sensitivity to detect analytes than an unbiased full scan MS analysis. The trade off for higher sensitivity is that novel metabolites cannot be detected, and all metabolites of interest must be accounted for prior to analysis.

Mass spectrometric analysis was performed on an ABI/Sciex 4000 Q-TRAP operating in scheduled multiple-reaction monitoring (sMRM) mode. Multiple-reaction monitoring (MRM) is a MS approach that monitors the transition of a parent ion to a specific daughter ion fragment. PGE2 and anandamide are used as examples to demonstrate the parent to daughter ion transition (Fig. 1). These defined parent and daughter ions are known as an MRM pair. An sMRM pair is an MRM pair with an associated liquid chromatography (LC) retention time. sMRM is an improvement over MRM allowing for better data collection and more analytes to be monitored in a single analysis. The MS operating software (Analyst 1.5) takes into account the total number of sMRM pairs and respective retention times, and optimizes how the mass spectrometer scans. We found that a 70 second retention time window for each sMRM pair was sufficient to account for metabolite peaks and slight shifts in their retention times. Although a feature on ABI/Sciex mass spectrometers running Analyst 1.5, scheduling MRM pairs can be replicated on triple quadrupole mass spectrometers from other companies. This method is intended as a starting point for the research community, which can be expanded upon as more metabolite standards become available, or scaled down to only include the metabolites of interest.

Figure 1.

Figure 1

The structures of the parent and daughter ions for PGE2 and AEA used in this method.

The switch from using MRM pairs to sMRM pairs has allowed us to increase the total number of metabolites monitored in our methodology, without sacrificing the quality of the data collected. Our previous eicosanoid methodology utilized manually dividing the entire mass spectrometric scan into 6 different 3-5 min periods, which contained about 20-30 metabolites with similar retention times. During each period, the mass spectrometer only monitored the contained subset of eicosanoids. This was done to maintain a duty cycle of less than 2 seconds. As more metabolites became available and added to the screen, it became a very laborious task to maintain an adequate duty cycle. Additionally, slight shifts in metabolite retention times due to column usage and variations in buffer preparations made it difficult to create and manage new periods. This methodology remained manageable as long as an upper limit of about 125 total MRM pairs was maintained. Our current eicosanoid methodology contains 171 total sMRM with improved sensitivity, where mass spectrometer algorithms have optimized the data collection process. These improvements are further discussed in the eicosanoid methodology and quantitation sections.

3.3. Eicosanoid methodology

We have developed a high-throughput CLASS approach to globally monitor a specific lipid class, the eicosanoids. Derived from poly-unsaturated fatty acids (PUFAs), all eicosanoids contain a conserved terminal carboxyl moiety (Fig. 1, PGE2). Eicosanoid are detected in negative electrospray ion mode to take advantage of the terminal carboxyl moiety, which is easily deprotonated during ionization. This feature allows eicosanoids to be detected without any additional derivatization. Unique sMRM pairs have been selected for each eicosanoid species. This was accomplished by analyzing product ion scans (MS/MS) from commercial eicosanoid standards (Cayman Chemical & Biomol). Additionally, optimal declustering potential (DP) and the collision energy (CE) parameters have been determined for each sMRM pair. These values were determined by directly infusing commercial standards into the mass spectrometer, while individually ramping these parameters in a MRM experiment. We have compiled the sMRM pair, DP, and CE values for each eicosanoid (Table 1). Table 1 also includes the biosynthetic pathway and PUFA that each eicosanoid is derived from.

Table 1. Optimized sMRM pairs and parameters for eicosanoids.

Fatty acid Pathway COMMON NAME Abbreviation Parent Daughter Retention Time (min)b LOD (pg) Declustering Potential Collision Energy Internal Standard
AA COX a(d4) 6-keto-Prostaglandin F1a (d4) 6k-PGF1a 373 167 5.0 ND -65 -35 -
AA COX (d4) Thromboxane B2 (d4) TXB2 373 173 6.3 ND -50 -25 -
AA COX (d4) Prostaglandin F2a (d4) PGF2a 357 197 6.9 ND -50 -30 -
AA COX (d4) Prostaglandin E2 (d4) PGE2 355 275 7.1 ND -50 -25 -
AA COX (d4) Prostaglandin D2 (d4) PGD2 355 275 7.5 ND -50 -25 -
AA COX (d4) 15-deoxy-Prostaglandin J2 (d4) 15d-PGJ2 319 275 14.5 ND -60 -20 -
AA COX (d4) 13,14-dihydro-15-keto Prostaglandin F2a (d4) dhk PGF2a 357 295 8.3 ND -80 -30 -
AA COX (d4) 13,14-dihydro-15-keto Prostaglandin E2 (d4) dhk PGE2 355 211 8.0 ND -45 -30 -
AA COX (d4) 13,14-dihydro-15-keto Prostaglandin D2 (d4) dhk PGD2 355 211 8.9 ND -45 -30 -
AA non-enz (d11) Isoprostane F-IV (d11) 5-iso PGF2a VI 364 115 6.6 ND -60 -30 -
AA LOX (d4) Leukotriene B4 (d4) LTB4 339 197 11.6 ND -70 -22 -
AA LOX (d8) 5-hydroxy-eicosatrienoic acid (d8) 5-HETE 327 116 16.7 ND -50 -20 -
AA LOX (d8) 12-hydroxy-eicosatrienoic acid (d8) 12-HETE 327 183 16.1 ND -60 -20 -
AA LOX (d8) 15-hydroxy-eicosatrienoic acid (d8) 15-HETE 327 226 15.5 ND -60 -20 -
AA CYP (d6) 20-hydroxy-eicosatrienoic acid (d6) 20-HETE 325 295 14.7 ND -70 -20 -
LA LOX (d4) 9-hydroxy-octadecadienoic acid (d4) 9-HODE 299 172 15.6 ND -60 -25 -
LA LOX (d4) 13-hydroxy-octadecadienoic acid (d4) 13-HODE 299 198 15.4 ND -60 -25 -
AA LOX (d7) 5-oxo-eicosatetraenoic acid (d7) 5-oxoETE 323 209 17.2 ND -50 -25 -
AA LOX (d4) Resolvin E1 (d4) RvE1 353 197 5.2 ND -70 -20 -
AA CYP (d8) 5(6)-epoxy-eicosatrienoic acid (d8) 5,6 EET 330 202 17.6 ND -30 -20 -
AA CYP (d8) 8(9)-epoxy-eicosatrienoic acid (d8) 8,9 EET 327 158 17.5 ND -60 -20 -
AA CYP (d8) 11(12)-epoxy-eicosatrienoic acid (d8) 11,12 EET 327 171 17.5 ND -60 -20 -
AA CYP (d8) 14(15)-epoxy-eicosatrienoic acid (d8) 14,15 EET 327 226 16.9 ND -50 -15 -
LA CYP (d4) 9,10-dihydroxy-octadecenoic acid (d4) 9,10 diHOME 317 203 13.0 ND -60 -30 -
LA CYP (d4) 12,13-dihydroxy-octadecenoic acid (d4) 12,13 diHOME 317 185 12.4 ND -60 -30 -
AA LOX (d5) Leukotriene C4 (d5) LTC4 630 272 9.2 ND -70 -35 -
AA LOX (d4) Leukotriene E4 (d4) LTE4 441 336 10.2 ND -60 -25 -
- - (d8) Arachidonic acid (d8) AA 311 267 20.6 ND -80 -20 -
- - (d5) Eicosapentaenoic acid (d5) EPA 306 262 19.3 ND -65 -15 -
- - (d5) Dohexacosaenoic acid (d5) DHA 332 234 20.2 ND -95 -20 -
AA COX 6-keto-Prostaglandin F 6k-PGF 369 163 5.1 1 -65 -35 (d4) 6k-PGF1α
AA COX Thromboxane B2 TXB2 369 169 6.3 1 -50 -25 (d4) TXB2
AA COX Prostaglandin F PGF 353 193 6.9 1 -50 -30 (d4) PGF
AA COX Prostaglandin E2 PGE2 351 271 7.1 1 -50 -25 (d4) PGE2
AA COX Prostaglandin D2 PGD2 351 271 7.5 1 -50 -25 (d4) PGD2
AA COX 11-beta-Prostaglandin F 11β PGF 353 193 6.2 ND -50 -30 (d4) PGF
DgLA COX Thromboxane B1 TXB1 371 171 6.1 ND -50 -25 (d4) TXB2
DgLA COX Prostaglandin F PGF 355 293 7.0 ND -75 -30 (d4) PGF
DgLA COX Prostaglandin E1 PGE1 353 273 7.4 ND -55 -25 (d4) PGE2
DgLA COX Prostaglandin D1 PGD1 353 273 7.5 ND -55 -25 (d4) PGD2
EPA COX Δ17-6-keto-Prostaglandin F Δ17 6k-PGF 367 163 4.4 ND -90 -35 (d4) PGF
EPA COX Thromboxane B3 TXB3 367 169 5.3 ND -50 -25 (d4) TXB2
EPA COX Prostaglandin F PGF 351 193 5.8 ND -75 -30 (d4) PGF
EPA COX Prostaglandin E3 PGE3 349 269 6.2 ND -55 -25 (d4) PGE2
EPA COX Prostaglandin D3 PGD3 349 269 6.5 ND -55 -25 (d4) PGD2
ADA COX dihomo Prostaglandin F dihomo PGF 381 221 8.9 ND -75 -35 (d4) PGF
ADA COX dihomo Prostaglandin E2 dihomo PGE2 379 299 9.1 ND -65 -30 (d4) PGE2
ADA COX dihomo Prostaglandin D2 dihomo PGD2 379 299 9.4 ND -65 -30 (d4) PGD2
ADA COX dihomo Prostaglandin J2 dihomo PGJ2 361 299 12.3 ND -55 -25 (d4) 15d-PGJ2
ADA COX dihomo 15-deoxy-Prostaglandin J2 dihomo 15d PGJ2 361 299 13.9 ND -55 -25 (d4) 15d-PGJ2
DgLA COX 6-keto-Prostaglandin E1 6k PGE1 367 143 5.3 ND -40 -25 (d4) PGE2
DgLA COX 6,15-diketo-, 13,14-dihydro-Prostaglandin F 6,15 dk-,dh-PGF 369 113 6.2 ND -60 -40 (d4) PGF
DgLA COX 15-keto-Prostaglandin F 15k PGF 353 113 7.4 ND -50 -35 (d4) PGF
AA COX 15-keto-Prostaglandin F 15k PGF 351 113 7.4 ND -40 -35 (d4) PGF
AA COX 15-keto-Prostaglandin E2 15k PGE2 349 113 7.5 1 -35 -30 (d4) PGE2
AA COX 13,14-dihydro-Prostaglandin F dh PGF 355 275 7.7 ND -40 -25 (d4) PGF
AA COX 13,14-dihydro-15-keto Prostaglandin F dhk PGF 353 291 8.1 1 -60 -25 (d4) dhk PGF
AA COX 13,14-dihydro-15-keto Prostaglandin E2 dhk PGE2 351 207 8.2 1 -40 -25 (d4) dhk PGE2
AA COX 13,14-dihydro-15-keto Prostaglandin D2 dhk PGD2 351 207 8.9 1 -40 -25 (d4) dhk PGD2
AA COX bicyclo Prostaglandin E2 bicyclo PGE2 333 113 10.6 ND -60 -35 (d4) PGE2
AA COX 11beta-13,14-dihydro-15-keto-Prostaglandin F 11β dhk PGF 353 113 8.0 ND -50 -35 (d4) PGF
AA COX 19-hydroxy-PGF 19oh PGF 369 193 3.3 ND -50 -35 (d4) PGF
AA COX 20-hydroxy-PGF 20oh PGF 369 193 3.2 ND -50 -35 (d4) PGF
AA COX 19-hydroxy-PGE2 19oh PGE2 367 189 3.6 ND -40 -25 (d4) PGE2
AA COX 20-hydroxy-PGE2 20oh PGE2 367 189 3.5 ND -40 -25 (d4) PGE2
AA COX 2,3-dinor-11-beta-Prostaglandin F 2,3 dinor 11β PGF 325 145 5.2 ND -40 -25 (d4) PGF
AA COX tetranor-Prostanglin F Metabolite tetranor-PGFM 329 293 2.5 ND -40 -25 (d4) PGF
AA COX tetranor-Prostaglandin E Metabolite tetranor-PGEM 327 291 2.5 ND -40 -25 (d4) PGE2
AA LOX tetranor 12-hydroxy-eicosatetraenoic acid tetranor 12-HETE 265 109 13.0 0.1 -75 -14 (d8) 15-HETE
AA COX 11-beta-Prostaglandin E2 11β PGE2 351 271 7.1 ND -40 -25 (d4) PGE2
AA COX Prostaglandin K2 PGK2 349 205 7.1 ND -50 -30 (d4) PGE2
AA COX 12S-hydroxy-heptadecatrienoic acid 12-HHT 279 163 13.4 1 -30 -30 (d8) 5-HETE
AA COX 11-hydroxy-eicosatetraenoic acid 11-HETE 319 167 16.0 0.1 -60 -20 (d8) 5-HETE
AA COX 11-hydroxy-eicosapentaenoic acid 11-HEPE 317 121 14.8 1 -70 -24 (d8) 15-HETE
AA COX 13-hydroxy-docosahexaenoic Acid 13-HDoHE 343 221 15.8 0.1 -60 -17 (d8) 15-HETE
AA COX Prostaglandin A2 PGA2 333 271 9.7 ND -30 -20 (d4) 15d-PGJ2
AA COX Prostaglandin B2 PGB2 333 271 9.5 ND -30 -20 (d4) 15d-PGJ2
AA COX 15-deoxy-Prostaglandin A2 15d-PGA2 315 271 15.0 ND -50 -15 (d4) 15d-PGJ2
AA COX Prostaglandin J2 PGJ2 333 271 9.7 1 -30 -20 (d4) 15d-PGJ2
AA COX 15-deoxy-Δ12,14-PGD2 15d-PGD2 333 271 11.7 1 -30 -20 (d4) 15d-PGJ2
AA COX 15-deoxy-Δ12,14-PGJ2 15d-PGJ2 315 271 14.5 0.1 -50 -15 (d4) 15d-PGJ2
AA non-enz Isoprostane F-IV 5-iso PGF VI 353 115 6.7 1 -60 -30 (d11) 5-iso PGF VI
AA non-enz Isoprostane F-III 8-iso PGF III 353 193 6.2 1 -50 -30 (d11) 5-iso PGF VI
AA non-enz 9-hydroxy-eicosatetraenoic acid 9-HETE 319 151 16.5 0.1 -60 -20 (d8) 5-HETE
EPA non-enz 9-hydroxy-eicosapentaenoic acid 9-HEPE 317 149 15.0 0.1 -75 -20 (d8) 5-HETE
DHA non-enz 8-hydroxy-docosahexaenoic Acid 8-HDoHE 343 109 16.2 0.1 -70 -20 (d8) 5-HETE
DHA non-enz 16-hydroxy-docosahexaenoic Acid 16-HDoHE 343 233 15.5 0.1 -75 -19 (d8) 15-HETE
DHA non-enz 20-hydroxy-docosahexaenoic Acid 20-HDoHE 343 241 15.3 0.1 -60 -20 (d8) 15-HETE
AA LOX Leukotriene B4 LTB4 335 195 11.5 1 -70 -22 (d4) LTB4
AA LOX 20-hydroxy-Leukotriene B4 20oh LTB4 351 195 5.1 ND -60 -25 (d4) LTB4
AA LOX 20-carboxy-Leukotriene B4 20cooh LTB4 365 195 5.1 ND -60 -25 (d4) LTB4
AA LOX 5,6 dihydroxy-eicosatetraenoic acid 5,6 diHETE 335 163 15.0 1 -60 -25 (d4) LTB4
AA LOX 6-trans-Leukotriene B4 6t LTB4 335 195 11.1 ND -70 -22 (d4) LTB4
AA LOX 12-epi-Leukotriene B4 12epi LTB4 335 195 11.1 ND -70 -22 (d4) LTB4
AA LOX 6-trans-,12-epi-Leukotnene B4 6t,12epi LTB4 335 195 11.1 ND -70 -22 (d4) LTB4
AA LOX 12-oxo-Leukotriene B4 12-oxoLTB4 333 179 12.4 ND -60 -17 (d4) LTB4
AA LOX Leukotriene C4 LTC4 625 272 9.2 ND -70 -28 (d5) LTC4
AA LOX Leukotriene D4 LTD4 495 177 10.6 ND -60 -25 (d4) LTE4
AA LOX Leukotriene E4 LTE4 438 333 10.2 1 -60 -25 (d4) LTE4
AA LOX 11-trans-Leukotriene C4 11t LTC4 625 272 9.8 ND -70 -35 (d5) LTC4
AA LOX 11-trans-Leukotriene D4 11t LTD4 495 177 11.5 ND -60 -25 (d4) LTE4
AA LOX 11-trans-Leukotriene E4 11t LTE4 438 333 10.9 ND -60 -25 (d4) LTE4
AA LOX 5-hydroxy-eicosatetraenoic acid 5-HETE 319 115 16.6 0.1 -60 -20 (d8) 5-HETE
EPA LOX 5-hydroxy-eicosapentaenoic acid 5-HEPE 317 115 15.3 0.1 -40 -17 (d8) 5-HETE
DHA LOX 7-hydroxy-docosahexaenoic acid 7-HDoHE 343 141 16.2 0.1 -60 -18 (d8) 5-HETE
DHA LOX 4-hydroxy-docosahexaenoic acid 4-HDoHE 343 101 16.8 0.1 -70 -17 (d8) 5-HETE
LA LOX 9-hydroxy-octadecatrienoic acid 9-HOTrE 293 171 14.1 0.1 -70 -20 (d8) 5-HETE
MA LOX 5-hydroxy-eicosatrienoic acid 5-HETrE 321 115 18.3 0.1 -70 -19 (d8) 5-HETE
AA LOX 5S,15S-dihydroxy-eicosatetraenoic acid 5,15 diHETE 335 201 11.1 1 -50 -30 (d4) LTB4
AA LOX 5(S),6(R)-Lipoxin A4 6R-LXA4 351 115 8.0 1 -30 -20 (d4) LTB4
AA LOX 5(S),6(S)-Lipoxin A4 6S-LXA4 351 115 8.3 1 -30 -20 (d4) LTB4
AA LOX 5(S),14(R)-Lipoxin A4 14R-LXA4 351 115 8.1 ND -70 -25 (d4) LTB4
EPA LOX Lipoxin A5 LXA5 349 113 6.7 ND -60 -25 (d4) LTB4
AA LOX Lipoxin B4 LXB4 351 221 7.3 ND -80 -25 (d4) LTB4
EPA LOX Resolvin E1 RvE1 349 195 5.2 ND -70 -20 (d4) RvE1
EPA LOX Resolvin D1 RvD1 375 141 7.8 ND -45 -20 (d4) LTB4
DHA LOX Neuroprotectin D1 PD1 359 153 10.5 ND -40 -20 (d4) LTB4
DHA LOX 15-trans Neuroprotectin D1 15t PD1 359 153 10.8 ND -40 -20 (d4) LTB4
DHA LOX 10S,17S-dihydroxy Docosahexaenoic acid 10S,17S-DiHDoHE 359 153 11.0 ND -40 -20 (d4) LTB4
AA LOX 8S,15S-dihydroxy-eicosatetraenoic acid 8,15 diHETE 335 127 10.7 1 -50 -25 (d4) LTB4
AA LOX 15-hydroxy-eicosatetraenoic acid 15-HETE 319 219 15.6 0.1 -50 -15 (d8) 15-HETE
EPA LOX 15-hydroxy-eicosapentaenoic acid 15-HEPE 317 219 14.6 0.1 -60 -20 (d8) 5-HETE
DHA LOX 17-hydroxy Docosahexaenoic acid 17-HDoHE 343 245 15.6 0.1 -60 -20 (d8)15-HETE
LA LOX 13-hydroxy-octadecadienoic acid 13-HODE 295 195 15.4 1 -60 -25 (d4) 13-HODE
aLA LOX 13-hydroxy-octadecatrienoic acid 13-HOTrE 293 195 14.3 1 -80 -25 (d4) 13-HODE
gLA LOX 13-hydroxy-g-octadecatrienoic acid 13-HOTre-g 293 193 14.5 0.1 -70 -20 (d4) 13-HODE
DgLA LOX 15-hydroxy-eicosatrienoic acid 15-HETrE 321 221 16.3 0.1 -70 -21 (d8) 15-HETE
AA LOX 8-hydroxy-eicosatetraenoic acid 8-HETE 319 155 16.3 0.1 -60 -20 (d8) 5-HETE
EPA LOX 8-hydroxy-eicosapentaenoic acid 8-HEPE 317 127 14.9 1 -70 -25 (d8) 5-HETE
DHA LOX 10-hydroxy Docosahexaenoic acid 10-HDoHE 343 181 15.8 1 -60 -17 (d8) 5-HETE
DgLA LOX 8-hydroxy-eicosatrienoic acid 8-HEtrE 321 157 16.8 1 -70 -23 (d8) 5-HETE
AA LOX Eoxin C4 EXC4 625 272 7.2 ND -70 -35 (d5) LTC4
AA LOX Eoxin D4 EXD4 495 177 10.7 ND -60 -25 (d4) LTE4
AA LOX Eoxin E4 EXE4 438 333 8.7 ND -60 -25 (d4) LTE4
AA LOX 12-hydroxy-eicosatetraenoic acid 12-HETE 319 179 16.2 1 -60 -20 (d8) 15-HETE
EPA LOX 12-hydroxy-eicosapentaenoic acid 12-HEPE 317 179 14.9 0.1 -70 -20 (d8) 15-HETE
DHA LOX 14-hydroxy-docosahexaenoic acid 14-HDoHE 343 205 15.8 1 -60 -18 (d8) 15-HETE
DHA LOX 11-hydroxy-docosahexaenoic acid 11-HDoHE 343 149 16.0 1 -60 -19 (d8) 5-HETE
LA LOX 9-hydroxy-octadecadienoic acid 9-HODE 295 171 15.6 1 -60 -25 (d4) 9-HODE
AA LOX Hepoxilin A3 HXA3 335 127 14.3 1 -50 -25 (d8) 14,15 EET
AA LOX Hepoxilin B3 HXB3 335 183 14.4 1 -40 -20 (d8) 14,15 EET
AA LOX 5-oxo-eicosatetraenoic acid 5-oxoETE 317 203 17.2 1 -40 -25 (d7) 5-oxoETE
AA LOX 12-oxo-eicosatetraenoic acid 12-oxoETE 317 153 16.1 1 -40 -25 (d7) 5-oxoETE
AA LOX 15-oxo-eicosatetraenoic acid 15-oxoETE 317 113 15.9 1 -40 -25 (d7) 5-oxoETE
LA LOX 9-oxo-octadecadienoic acid 9-oxoODE 293 185 16.1 1 -60 -25 (d7) 5-oxoETE
LA LOX 13-oxo-octadecadienoic acid 13-oxoODE 293 113 15.7 1 -70 -30 (d7) 5-oxoETE
EPA LOX 15-oxo-eicosadienoic acid 15-oxoEDE 321 113 17.7 1 -100 -32 (d7) 5-oxoETE
AA CYP 20-hydroxy-eicosatetraenoic acid 20-HETE 319 289 14.8 1 -75 -25 (d6) 20-HETE
AA CYP 19-hydroxy-eicosatetraenoic acid 19-HETE 319 231 14.6 1 -80 -25 (d6) 20-HETE
AA CYP 18-hydroxy-eicosatetraenoic acid 18-HETE 319 261 15.0 0.1 -80 -25 (d6) 20-HETE
AA CYP 17-hydroxy-eicosatetraenoic acid 17-HETE 319 247 15.1 0.1 -80 -25 (d6) 20-HETE
AA CYP 16-hydroxy-eicosatetraenoic acid 16-HETE 319 189 15.1 1 -80 -25 (d6) 20-HETE
EPA CYP 18-hydroxy-eicosapentaenoic acid 18-HEPE 317 215 14.2 0.1 -60 -20 (d6) 20-HETE
AA CYP 5(6)-epoxy-eicosathenoic acid 5,6 EET 319 191 17.8 1 -30 -20 (d8) 5,6 EET
AA CYP 8(9)-epoxy-eicosatrienoic acid 8,9 EET 319 155 17.5 1 -60 -20 (d8) 8,9 EET
AA CYP 11(12)-epoxy-eicosatrienoic acid 11,12 EET 319 167 17.3 1 -60 -20 (d8) 11,12 EET
AA CYP 14(15)-epoxy-eicosatrienoic acid 14,15 EET 319 219 16.9 1 -50 -15 (d8) 14,15 EET
EPA CYP 14,15-epoxy Eicosatetraenoic acid 14,15 EpETE 317 208 15.8 1 -65 -18 (d8) 14,15 EET
EPA CYP 17,18-epoxy Eicosatetraenoic acid 17,18 EpETE 317 215 15.6 1 -65 -16 (d8) 14,15 EET
DHA CYP 16,17-epoxy Docosapentaenoic acid 16,17 EpDPE 343 193 17.0 1 -70 -16 (d8) 14,15 EET
DHA CYP 19,20-epoxy Docosapentaenoic acid 19,20 EpDPE 343 241 16.5 1 -70 -17 (d8) 14,15 EET
DHA CYP 19,20-dihydroxy-docosapentaenoic acid 19,20 DiHDPA 361 229 13.1 1 -70 -25 (d8) 14,15 EET
LA CYP 9(10)-epoxy-octadecenoic acid 9,10 EpOME 295 171 17.1 1 -60 -25 (d4) 9,10 diHOME
LA CYP 12(13)-epoxy-octadecenoic acid 12,13 EpOME 295 195 17.1 1 -60 -25 (d4) 12,13 diHOME
AA CYP 5,6-dihydroxy-eicosatrienoic acid 5,6 DHET 337 145 15.0 0.1 -75 -25 (d8) 5-HETE
AA CYP 8,9-dihydroxy-eicosatrienoic acid 8,9 DHET 337 127 14.4 1 -60 -30 (d8) 5-HETE
AA CYP 11,12-dihydroxy-eicosatrienoic acid 11,12 DHET 337 167 14.0 0.1 -60 -25 (d8) 5-HETE
AA CYP 14,15-dihydroxy-eicosatrienoic acid 14,15 DHET 337 207 13.3 0.1 -60 -25 (d8) 5-HETE
LA CYP 9,10-dihydroxy-octadecenoic acid 9,10 diHOME 313 201 12.9 0.1 -60 -30 (d4) 9,10 diHOME
LA CYP 12,13-dihydroxy-octadecenoic acid 12,13 diHOME 313 183 12.4 1 -60 -30 (d4) 12,13 diHOME
- - Arachidonic acid AA 303 259 20.6 ND -80 -20 (d8) AA
- - Adrenic acid ADA 331 287 21.3 ND -80 -20 (d8) AA
- - Eicosapentaenoic acid EPA 301 257 19.4 ND -65 -15 (d5) EPA
- - Docohexaenoic acid DHA 327 229 20.2 ND -95 -20 (d5) DHA
a

deturated internal standards are shaded in grey.

b

rentention time are representative values.

Metabolite identification is achieved by combining both ESI-MS/MS and HPLC analyses. The detection of an sMRM pair with an associated retention time from a reverse-phase HPLC gradient for a particular lipid metabolite is matched to values obtained from a pure commercial standard subjected to the same LC/MS/MS analysis. Since chromatography fluctuates with column usage and variations in buffer preparation, it is imperative that eicosanoid standards are analyzed as part of every experiment. Our methodology contains 171 sMRM pairs (141 metabolites + 30 deuterated internal standards) for eicosanoids and related metabolites, which are monitored in a single 25 min LC/MS/MS analysis. Representative retention time values for each eicosanoid are reported in Table 1. Representative retention times are reported because retention fluctuations occur between experiments which can be due to column usage, buffer preparation, and instrument variation. A chromatograph from a single analysis of a 100 ng primary standard solution containing a total of 102 eicosanoid species demonstrates the robustness of our methodology (Fig. 2A). Specific eicosanoid ion chromatographs have been extracted to illustrate different features of our methodology (Fig. 2B). Chromatographic separation is paramount when monitoring a large number of structurally similar metabolites. The COX pathway derived metabolites, prostaglandin E2 (PGE2) and prostaglandin D2 (PGD2) have the same molecular formula, differing only by the placement of a keto- and hydroxyl- group at position 9 and 11. Subsequent full MS/MS scan analyses of these two molecules produce the exact same ion fragmentation pattern. The use of mass spectrometry alone is not sufficient to distinguish between metabolites that share the same sMRM pair. Adequate chromatographic separation of PGE2 and PGD2 allows us to differentiate between these two metabolites (Fig. 2B). There are 39 occurrences in our eicosanoid methodology where a metabolite shares an sMRM pair with another metabolite.

Figure 2.

Figure 2

Chromatograph from a single 100 ng standard solution subjected to our eicosanoid methodology. (A) 102 sMRM pairs were extracted from a single run. (B) Selected eicosanoid sMRM pairs from the major biosynthetic pathways were extracted from a single analysis. (C) A magnified view of the chromatograph of extracted HETE and HODE metabolites.

Adequate chromatographic separation is not always achieved, due to the high amount of structural similarity between eicosanoid species. Many eicosanoids have slight modifications to their molecular structure, while maintaining the exact molecular weight. This is exemplified in the lipoxygenase generated HETE (20-carbon) and HODE (18-carbon) metabolites that differ at the position of a single hydroxyl group. Our HPLC gradient produces only minimal chromatographic separation of these metabolites, leading to overlap of their elution profiles (Fig. 2C). These metabolites produce many of the same daughter ion fragments, making it difficult to distinguish each eicosanoid species using a full MS/MS scan. Our methodology overcomes this obstacle by utilizing a unique sMRM pair for each of these lipid metabolites. Our sMRM approach is able to detect which metabolites are present, even when a cluster of metabolites display overlapping elution profiles (Fig. 2C).

3.4. N-acylethanolamine (NAE) methodology

A similar CLASS approach used to monitor eicosanoids is employed in the detection of NAEs. The NAE methodology can be employed as a lone analysis or performed in tandem after samples have subjected to the eicosanoid methodology. All NAEs contain an acyl linkage to the nitrogen of an ethanolamine molecule (Fig. 1, AEA). NAEs are detected in positive electrospray ion mode. Unlike the eicosanoids, NAEs ionize more efficiently in positive ion mode due to their conserved amide nitrogen. Since the carboxyl-amide bond is labile, fragmentation of an N-acylethanolamine [M+H]+ species produces a 62 m/z ion (C2H8N1O1+). The presence of salts such as Na+ and K+, which can remain associated with these lipids even after sample preparation, can facilitate the formation of other NAE ion species such as [M+Na]+ and [M+K]+, diminishing the overall sensitivity of these lipids. Ammonium acetate, when used in concentrations above 1 mM, can shift the equilibrium toward [M+NH4]+ species, which degrade to [M+H]+ + NH3 during ionization. Similarly to the eicosanoids, no additional chemical derivatization is required to monitor NAEs. The current NAE methodology monitors 36 different metabolites, derived from saturated fatty acids, mono- to poly- unsaturated fatty acids, and eicosanoids. Some of these metabolites can be purchased (Cayman Chemical), while others were synthesized in our laboratory by standard methods. The sMRM pair and representative retention time for each NAE have been determined in the same manner as the eicosanoids (Table 2). Fragmentation of the carboxyl-amide bond required the same declustering potential (50) and collision energy (40) values for all NAEs.

Table 2. Optimized sMRM pairs and parameters for N-acylethanolamines.

COMMON NAME Abbreviation Parent Daughter Retention Time (min)b LOD (pg) Internal Standard
a(d4) Prostaglandin F Ethanolamide (d4) PGF-EA 384 62 5.1 ND -
(d4) Palmitoyl Ethanolamide (d4) 16:0-EA 312 62 12.1 ND -
(d4) Oleoyl Ethanolamide (d4) 18:1-EA 328 62 12.5 ND -
(d8) Arachidonyl Ethanolamide (d8) 20:4-EA 356 62 11.4 ND -
Prostaglandin F Ethanolamide PGF-EA 380 62 5.1 1,000 (d4) PGF-EA
11-beta-Prostaglandin F Ethanolamide 11β PGF-EA 380 62 4.8 ND (d4) PGF-EA
8-iso-Prostaglandin F III Ethanolamide 8-iso-PGF III-EA 380 62 4.7 100 (d4) PGF-EA
Prostaglandin E2 Ethanolamide PGE2-EA 378 62 5.2 100 (d4) PGF-EA
Prostaglandin D2 Ethanolamide PGD2-EA 378 62 5.4 10 (d4) PGF-EA
5(6)-Epoxy-Eicosatrienoic acid Ethanolamide 5,6-EET-EA 346 62 9.8 1 (d4) PGF-EA
8(9)-Epoxy-Eicosatrienoic acid Ethanolamide 8,9-EET-EA 346 62 9.4 10 (d4) PGF-EA
11(12)-Epoxy-Eicosatrienoic acid Ethanolamide 11,12-EET-EA 346 62 9.2 1 (d4) PGF-EA
14(15)-Epoxy-Eicosatrienoic acid Ethanolamide 14,15-EET-EA 346 62 8.9 1 (d4) PGF-EA
15-Hydroxy-Eicosatetraenoic acid Ethanolamide 15-HETE-EA 346 62 8.4 0.1 (d4) PGF-EA
20-Hydroxy-Eicosatetraenoic acid Ethanolamide 20-HETE-EA 346 62 7.6 0.1 (d4) PGF-EA
Lauroyl Ethanolamide 12:0-EA 244 62 8.9 10 (d4) 16:0-EA
Myristoyl Ethanolamide 14:0-EA 272 62 10.6 10 (d4) 16:0-EA
Pentadecanoyl Ethanolamide 15:0-EA 286 62 11.4 0.1 (d4) 16:0-EA
Palmitoyl Ethanolamide 16:0-EA 300 62 12.2 0.1 (d4) 16:0-EA
Heptadecanoyl Ethanolamide 17:0-EA 314 62 12.9 0.1 (d4) 16:0-EA
Stearoyl Ethanolamide 18:0-EA 328 62 13.7 0.1 (d4) 16:0-EA
Arachidoyl Ethanolamide 20:0-EA 356 62 14.8 0.1 (d4) 16:0-EA
Tricosanoyl Ethanolamide 23:0-EA 398 62 16.2 0.1 (d4) 16:0-EA
Lignoceroyl Ethanolamide 24:0-EA 412 62 16.6 0.1 (d4) 16:0-EA
Palmitoleoyl Ethanolamide 16:1-EA 298 62 11.1 0.1 (d4) 18:1-EA
Oleoyl Ethanolamide 18:1-EA 326 62 12.5 0.1 (d4) 18:1-EA
Docosaenoyl Ethanolamide 22:1-EA 382 62 14.9 0.1 (d4) 18:1-EA
Nervonoyll Ethanolamide 24:1-EA 410 62 15.8 0.1 (d4) 18:1-EA
Linoleoyl Ethanolamide 18:2-EA 324 62 11.5 0.1 (d8) 20:4-EA
α-Linolenoyl Ethanolamide 18:3(α)-EA 322 62 10.8 10 (d8) 20:4-EA
γ-Linolenoyl Ethanolamide 18:3(γ)-EA 322 62 10.4 100 (d8) 20:4-EA
Meadoyl Ethanolamide 20:3-EA 350 62 12.4 0.1 (d8) 20:4-EA
Dihomo-γ-Linolenoyl Ethanolamide 20:3-EA (DγLA) 350 62 12 0.1 (d8) 20:4-EA
Arachidonoyl Ethanolamide 20:4-EA 348 62 11.4 0.1 (d8) 20:4-EA
Docosatetraenoyl Ethanolamide 22:4-EA 376 62 12.5 0.1 (d8) 20:4-EA
Eicosapenaenoyl Ethanolamide 20:5-EA 346 62 10.5 0.1 (d8) 20:4-EA
Docohexaenoyl Ethanolamide 22:6-EA 372 62 11.2 0.1 (d8) 20:4-EA
a

deturated internal standards are shaded in grey.

b

rentention time are representative values.

The NAE metabolites are identified using a 25 min LC/MS/MS analysis. The identity of detected NAE metabolites are confirmed against pure NAE standards subjected to the same LC/MS/MS analysis. A chromatograph from a 100 ng standard stock solution containing 33 NAEs subjected to this methodology is depicted in Fig. 3A. Selected metabolites have been extracted to simplify the data produced by this method (Fig. 3B). Since every NAE generates the daughter ion fragment (62 m/z)+, many of metabolites share the same MRM transition. This can be seen when examining the different HETE-EA and EET-EA species which all have a 346.0/62.0 sMRM transition (Fig. 3C). Having adequate chromatographic separation is key in distinguishing these metabolites from each other. There are 15 NAEs that have a shared sMRM pair.

Figure 3.

Figure 3

Chromatograph from a single 100 ng standard solution subjected to our ethanolamine methodology. (A) 40 sMRM pairs were extracted from a single run. (B) A diverse group of selected ethanolamides sMRM pairs were extracted from a single analysis. (C) A magnified view of the chromatograph of extracted HETE-EA and EET-EA metabolites.

3.5. Quantitation

Our current methodology allows for both relative and exact quantitation using a stable isotope dilution technique [35]. This technique applies for the quantitation of eicosanoids and NAEs. First, we employ measures to filter out aberrant signals from the dataset, whereby a detected peak is considered valid if the peak's height is 3-fold greater than the background noise. Peaks that do not meet this requirement are disregarded. Also, the addition of deuterated internal standards can add contaminating lipid metabolites to the sample, which arise from impurities in the standard. The deuterated internal standard is mass analyzed in every experiment, and contaminating peaks are subtracted from the samples.

Both relative and exact quantitation heavily relies upon making comparisons between the integrated areas of a given lipid metabolite and its corresponding internal standard (AreaMETABOLITE / AreaINSTD). The internal standard solution for the eicosanoids is comprised of 30 deuterated eicosanoids, while the NAE's internal standard solution contains 4 deuterated NAEs. The criteria used to pair a metabolite with a particular internal standard include: 1) matching the metabolite with the analogous deuterated species (PGD2 and d4-PGD2), or 2) matching the metabolite with an internal standard that has a similar chemical structure and retention time (PGJ2 and d4-15d-PGJ2). Table 1 and 2 lists the deuterated internal standards (highlighted in gray) and lipid metabolite they were paired with. This ratio, which is established prior to lipid extraction, is maintained throughout the entire sampling process. This technique allows for accurate quantitation because lipid metabolites and corresponding internal standards have similar ionization efficiencies and will be equally affected by factors such as ion suppression. An important caveat is that the internal standard must be detectable above the background for quantitation.

Relative quantitation is determined by comparing the ratio (AreaMETABOLITE / AreaINSTD) between two different samples. This can be useful in monitoring how a single or a group of lipid metabolites change during different stages of a disease, or how these metabolic pathways are globally affected by pharmacological intervention. To quantitate the exact amount of a given metabolite in a sample, a primary standard curve generated from commercially bought standards is required. Primary standard curves for eicosanoids and NAE are generated separately. A primary standard curve is produced from 7 different concentrated eicosanoid or NAE standard solutions (0.1 ng, 0.3 ng, 1 ng, 3 ng, 10 ng, 30 ng, and 100 ng) that have been spiked with the internal standard solution. Since the primary standard and internal standard solutions are maintained in 100% ethanol, the addition of aqueous buffer is required to reproduce a metabolite's chromatographic retention time. For eicosanoids, an equal volume of 0.2% acetic acid-water is added to the primary / internal standard mixture, while an equal volume of water-acetonitrile-acetic acid (70:30:0.1; v/v/v) + 1 g/L ammonium acetate is added to the NAE primary / internal standard mixture. Each primary standard concentration is analyzed in duplicate and averaged. The primary standard curve is determined by generating a linear regression trend line that is forced through 0.

Representative primary standard curves from eicosanoid and NAE metabolites are shown to illustrate this technique (Fig. 4). These primary standard curves were generated from 3 separate experiments performed in duplicate spanning a 2-month period. Each standard curve displays a correlation value (R2 value) above 0.999 indicating the high reproducibility of this methodology. Exact quantitation of a metabolite in a given sample is determined by extrapolating the amount (X-axis value) from where the ratio ((AreaMETABOLITE / AreaINSTD); (Y-axis value)) intercepts the primary standard curve (Fig. 4). Alternately, the exact amount of a given lipid metabolite in a sample can be determined by dividing the ratio (AreaMETABOLITE / AreaINSTD) by the slope of the standard curve. We routinely quantitate about 100 eicosanoids out of the total 141 monitored in a single analysis. Our NAE methodology contains 33 quantifiable metabolites out of a total 36 monitored in a single analysis. Some metabolites are not quantitated because either a pure standard is not available (such as the dihomo-prostaglandins) or are routinely observed in very low abundance in experimental models that we have examined

Figure 4.

Figure 4

Primary standard curves from representative eicosanoid and ethanolamine metabolites were generated from 3 separate experiments performed in duplicate. Primary standard curves were generated from 7 concentrations ranging from 0.1 – 100 ng. Error bars represent the standard error of the mean.

Monitoring sMRM pairs instead of full MS/MS scans greatly increases the sensitivity of detection. A 100 ng mixture of standards containing eicosanoids or NAE were serially diluted and analyzed by our methodology to determine the lower limit of detection (LOD) for a portion of the routinely quantitated eicosanoids and NAEs. These values are detailed in Table 1 and 2. The LOD for the eicosanoids analyzed ranged between 0.1 pg -1 pg. This is a definite improvement over our previous method with reported LOD values ranging from 1 pg to 10 pg on average [30]. The LOD values for a majority of NAEs were detected at 0.1 pg, while in a few instances a higher LOD value was observed ranging from 10 pg – 1000 pg. The difference in sensitivity is due to the efficiency and stability of the parent to daughter ion transition.

3.6. Application of the lipidomic methodology

The application of these methodologies can be used to globally monitor the changes in eicosanoid and NAE metabolites in tissue samples. As an example, cerebral spinal fluid (CSF) and lumbar spinal cord sections from rats injected with the hyperalgesia-inducing agent carrageenan in their hind paw were analyzed by both eicosanoid and NAE methodologies performed in series [19]. The version of eicosanoid methodology employed during this study monitored fewer eicosanoid species (124 sMRM). The data obtained from an extensive 24 h time-course experiment is graphed as a heat map to show the global representation of the relative changes of these metabolites (Fig. 5). The power of this approach can be seen in the emergent patterns from large complex data sets. The administration of peripheral carrageenan caused an increase in central levels of arachidonic acid-derived (AA) COX metabolites. Also, 12-LOX and corresponding dehydration metabolites were also observed to increase. There is an extensive amount of literature on the involvement of PGE2 (COX metabolite) in central pain signaling, however, little is known about the role that 12-LOX metabolites play in this process. These observations could have been missed if only a single or select group of lipid metabolites was monitored. The capability to globally monitor these lipids has led to the identification of new bioactive mediators.

Figure 5.

Figure 5

A time-course heat map of cerebral spinal fluid (CSF) and the ipsilateral of the portion lumbar spinal cord (IPSI) from rats treated with the hyperalgesia-inducing agent carrageenan. Metabolites are clustered based on their biosynthetic pathway. This data is adapted from Buczynski et al (16).

Distinct patterns are observed when comparing the eicosanoid and NAE profiles from the CSF and spinal cord samples. Fewer total lipid metabolites are detected in CSF samples, containing only a portion of the eicosanoids detected in spinal cord (Fig. 5). Also, NAEs are only detected in the spinal cord sample. Temporal patterns between lipid metabolites present in both samples are observed. Examination of quantitated amounts of PGE2 exemplifies the temporal differences between CSF and spinal cord (Fig. 6). PGE2 peaks at 4 h in CSF then returns to basal levels by 24 h. In contrast, PGE2 levels begin to rise at later times in the spinal cord and remain elevated. Additionally, the NAE anandamide (20:4-EA) is observed to significantly increase at later time points in the spinal cord (Fig. 6).

Figure 6.

Figure 6

Quantitative amounts of eicosanoid and ethanolamine in CSF and spinal cord over a 24 h time-course. Error bars represent the standard error of the mean. * indicate p-values < 0.05. This data is adapted from Buczynski et al (16).

4. Summary

We present a targeted CLASS approach to globally monitor and quantitate eicosanoids and N-acylethanolamines. Our current eicosanoid methodology represents a distinct advance over our previous versions, increasing the total number of sMRM pairs monitored from 60 to 171. The introduction of scheduled MRM (sMRM) pairs, has allowed for the detection of more metabolites, and the number is only limited by the number of available lipid metabolite standards. The increase in the total metabolites monitored has not compromised the quality of the data collection process. Software advancements have allowed for greater sensitivity and removed a lot of the tediousness of monitoring a very large set of metabolites. This methodology facilitates more thorough metabolite studies and can easily be adapted to other metabolite classes.

Research Highlights.

> We present a high-throughput Lipidomics methodology for eicosanoids and N-acylethanolamines. > The rationale behind the method design is addressed. > Quantitation for 100 eicosanoid and 33 N-acylethanolamine species. > Limits of detections are determined. > Biological application of the methodology is demonstrated.

Acknowledgments

This work was supported by the LIPID MAPS Large Scale Collaborative Grant GM069338 and by GM064611

The abbreviations used are

NAE

N-acylethanolamine

sMRM

scheduled multiple reaction monitoring

LIPID MAPS

LIPID Metabolites And Pathways Strategy

AA

arachidonic acid

20:4; AEA

ananamide

COX

cyclooxygenase

LOX

lipoxygenase

CYP

cytochrome P450

15d-PGJ2

15-deoxy-Δ12,14-PGJ2

PGE2

prostaglandin E2

PGD2

prostaglandin D2

PGJ2

prostaglandin J2

dhk-PGD2

13,14-dihydro-15-keto prostaglandin D2

5-iso PGF IV

5-iso-prostaglandin F IV

5,6 EET-EA

5,6-epoxy-eicosatetrienoic acid ethanolamide

8,9EET-EA

8,9-epoxy-eicosatetrienoic acid ethanolamide

11,12 EET-EA

11,12-epoxy-eicosatetrienoic acid ethanolamide

14,15 EET-EA

14,15-epoxy-eicosatetrienoic acid ethanolamide

15-HETE-EA

15-hydroxy-eicosapentaenoic acid ethanolamide

PGF-EA

prostaglandin F ethanolamide

20:3-EA

dihomo-γ-linolenoyl ethanolamide

16:0-EA

lauroyl ethanolamide

18:2-EA

linoleoyl ethanolamide

24:0-EA

lignoceryol ethanolamide

5-HETE

5-hydroxy-eicosapentaenoic acid

16-HETE

16-hydroxy-eicosapentaenoic acid

17-HETE

17-hydroxy-eicosapentaenoic acid

18-HETE

18-hydroxy-eicosapentaenoic acid

20-HETE

20-hydroxy-eicosapentaenoic acid

LTB4

Leukotriene B4

9-HODE

9-hydroxy-octadecatrienoic acid

13-HODE

13-hydroxy-octadecatrienoic acid

9,10-diHOME

9,10-dihydro-octadecenoic acid

12,13-diHOME

12,13-dihydro-octadecenoic acid

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Funk CD. Science. Vol. 294. New York, N.Y: 2001. Prostaglandins and leukotrienes: advances in eicosanoid biology; pp. 1871–1875. [DOI] [PubMed] [Google Scholar]
  • 2.Buczynski MW, Dumlao DS, Dennis EA. Thematic Review Series: Proteomics. An integrated omics analysis of eicosanoid biology. Journal of lipid research. 2009;50:1015–1038. doi: 10.1194/jlr.R900004-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Harkewicz R, D EA. Application of Mass Spectrometry to Lipids and Membranes. Annual Review Biochemistry. 2011 doi: 10.1146/annurev-biochem-060409-092612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Rodriguez-Antona C, Ingelman-Sundberg M. Cytochrome P450 pharmacogenetics and cancer. Oncogene. 2006;25:1679–1691. doi: 10.1038/sj.onc.1209377. [DOI] [PubMed] [Google Scholar]
  • 5.Hyde CA, Missailidis S. Inhibition of arachidonic acid metabolism and its implication on cell proliferation and tumour-angiogenesis. International immunopharmacology. 2009;9:701–715. doi: 10.1016/j.intimp.2009.02.003. [DOI] [PubMed] [Google Scholar]
  • 6.Weitz D, Weintraub H, Fisher E, Schwartzbard AZ. Fish oil for the treatment of cardiovascular disease. Cardiology in review. 18:258–263. doi: 10.1097/CRD.0b013e3181ea0de0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Calder PC, Zurier RB. Polyunsaturated fatty acids and rheumatoid arthritis. Current opinion in clinical nutrition and metabolic care. 2001;4:115–121. doi: 10.1097/00075197-200103000-00006. [DOI] [PubMed] [Google Scholar]
  • 8.Harmon GS, Dumlao DS, Ng DT, Barrett KE, Dennis EA, Dong H, Glass CK. Pharmacological correction of a defect in PPAR-gamma signaling ameliorates disease severity in Cftr-deficient mice. Nature medicine. 16:313–318. doi: 10.1038/nm.2101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Pierre SR, Lemmens MA, Figueiredo-Pereira ME. Subchronic infusion of the product of inflammation prostaglandin J2 models sporadic Parkinson's disease in mice. Journal of neuroinflammation. 2009;6:18. doi: 10.1186/1742-2094-6-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sala A, Folco G, Murphy RC. Transcellular biosynthesis of eicosanoids. Pharmacol Rep. 62:503–510. doi: 10.1016/s1734-1140(10)70306-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Six DA, Dennis EA. The expanding superfamily of phospholipase A(2) enzymes: classification and characterization. Biochimica et biophysica acta. 2000;1488:1–19. doi: 10.1016/s1388-1981(00)00105-0. [DOI] [PubMed] [Google Scholar]
  • 12.Schaloske RH, Dennis EA. The phospholipase A2 superfamily and its group numbering system. Biochimica et biophysica acta. 2006;1761:1246–1259. doi: 10.1016/j.bbalip.2006.07.011. [DOI] [PubMed] [Google Scholar]
  • 13.Nomura DK, Blankman JL, Simon GM, Fujioka K, Issa RS, Ward AM, Cravatt BF, Casida JE. Activation of the endocannabinoid system by organophosphorus nerve agents. Nature chemical biology. 2008;4:373–378. doi: 10.1038/nchembio.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Long JZ, Nomura DK, Vann RE, Walentiny DM, Booker L, Jin X, Burston JJ, Sim-Selley LJ, Lichtman AH, Wiley JL, Cravatt BF. Dual blockade of FAAH and MAGL identifies behavioral processes regulated by endocannabinoid crosstalk in vivo. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:20270–20275. doi: 10.1073/pnas.0909411106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Simmons DL, Botting RM, Hla T. Cyclooxygenase isozymes: the biology of prostaglandin synthesis and inhibition. Pharmacological reviews. 2004;56:387–437. doi: 10.1124/pr.56.3.3. [DOI] [PubMed] [Google Scholar]
  • 16.Sacerdoti D, Gatta A, McGiff JC. Role of cytochrome P450-dependent arachidonic acid metabolites in liver physiology and pathophysiology. Prostaglandins & other lipid mediators. 2003;72:51–71. doi: 10.1016/s1098-8823(03)00077-7. [DOI] [PubMed] [Google Scholar]
  • 17.Buczynski MW, Parsons LH. Quantification of brain endocannabinoid levels: methods, interpretations and pitfalls. British journal of pharmacology. 160:423–442. doi: 10.1111/j.1476-5381.2010.00787.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ueda N, Tsuboi K, Uyama T. Enzymological studies on the biosynthesis of N-acylethanolamines. Biochimica et biophysica acta. 1801:1274–1285. doi: 10.1016/j.bbalip.2010.08.010. [DOI] [PubMed] [Google Scholar]
  • 19.Buczynski MW, Svensson CI, Dumlao DS, Fitzsimmons BL, Shim JH, Scherbart TJ, Jacobsen FE, Hua XY, Yaksh TL, Dennis EA. Inflammatory hyperalgesia induces essential bioactive lipid production in the spinal cord. Journal of neurochemistry. 114:981–993. doi: 10.1111/j.1471-4159.2010.06815.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ahn K, Johnson DS, Mileni M, Beidler D, Long JZ, McKinney MK, Weerapana E, Sadagopan N, Liimatta M, Smith SE, Lazerwith S, Stiff C, Kamtekar S, Bhattacharya K, Zhang Y, Swaney S, Van Becelaere K, Stevens RC, Cravatt BF. Discovery and characterization of a highly selective FAAH inhibitor that reduces inflammatory pain. Chem Biol. 2009;16:411–420. doi: 10.1016/j.chembiol.2009.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Karbarz MJ, Luo L, Chang L, Tham CS, Palmer JA, Wilson SJ, Wennerholm ML, Brown SM, Scott BP, Apodaca RL, Keith JM, Wu J, Breitenbucher JG, Chaplan SR, Webb M. Biochemical and biological properties of 4-(3-phenyl-[1,2,4] thiadiazol-5-yl)-piperazine-1-carboxylic acid phenylamide, a mechanism-based inhibitor of fatty acid amide hydrolase. Anesth Analg. 2009;108:316–329. doi: 10.1213/ane.0b013e31818c7cbd. [DOI] [PubMed] [Google Scholar]
  • 22.Wang X, Sarris K, Kage K, Zhang D, Brown SP, Kolasa T, Surowy C, El Kouhen OF, Muchmore SW, Brioni JD, Stewart AO. Synthesis and evaluation of benzothiazole-based analogues as novel, potent, and selective fatty acid amide hydrolase inhibitors. J Med Chem. 2009;52:170–180. doi: 10.1021/jm801042a. [DOI] [PubMed] [Google Scholar]
  • 23.Piomelli D, Tarzia G, Duranti A, Tontini A, Mor M, Compton TR, Dasse O, Monaghan EP, Parrott JA, Putman D. Pharmacological profile of the selective FAAH inhibitor KDS-4103 (URB597) CNS Drug Rev. 2006;12:21–38. doi: 10.1111/j.1527-3458.2006.00021.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Reinke M. Monitoring thromboxane in body fluids: a specific ELISA for 11-dehydrothromboxane B2 using a monoclonal antibody. The American journal of physiology. 1992;262:E658–662. doi: 10.1152/ajpendo.1992.262.5.E658. [DOI] [PubMed] [Google Scholar]
  • 25.Shono F, Yokota K, Horie K, Yamamoto S, Yamashita K, Watanabe K, Miyazaki H. A heterologous enzyme immunoassay of prostaglandin E2 using a stable enzyme-labeled hapten mimic. Analytical biochemistry. 1988;168:284–291. doi: 10.1016/0003-2697(88)90320-x. [DOI] [PubMed] [Google Scholar]
  • 26.Baranowski R, Pacha K. Gas chromatographic determination of prostaglandins. Mini reviews in medicinal chemistry. 2002;2:135–144. doi: 10.2174/1389557024605465. [DOI] [PubMed] [Google Scholar]
  • 27.Murphy RC, Barkley RM, Zemski Berry K, Hankin J, Harrison K, Johnson C, Krank J, McAnoy A, Uhlson C, Zarini S. Electrospray ionization and tandem mass spectrometry of eicosanoids. Analytical biochemistry. 2005;346:1–42. doi: 10.1016/j.ab.2005.04.042. [DOI] [PubMed] [Google Scholar]
  • 28.Margalit A, Duffin KL, Isakson PC. Rapid quantitation of a large scope of eicosanoids in two models of inflammation: development of an electrospray and tandem mass spectrometry method and application to biological studies. Analytical biochemistry. 1996;235:73–81. doi: 10.1006/abio.1996.0093. [DOI] [PubMed] [Google Scholar]
  • 29.Quehenberger O, Armando A, Dumlao D, Stephens DL, Dennis EA. Lipidomics analysis of essential fatty acids in macrophages. Prostaglandins, leukotrienes, and essential fatty acids. 2008;79:123–129. doi: 10.1016/j.plefa.2008.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Deems R, Buczynski MW, Bowers-Gentry R, Harkewicz R, Dennis EA. Detection and quantitation of eicosanoids via high performance liquid chromatography-electrospray ionization-mass spectrometry. Methods in enzymology. 2007;432:59–82. doi: 10.1016/S0076-6879(07)32003-X. [DOI] [PubMed] [Google Scholar]
  • 31.Buczynski MW, Stephens DL, Bowers-Gentry RC, Grkovich A, Deems RA, Dennis EA. TLR-4 and sustained calcium agonists synergistically produce eicosanoids independent of protein synthesis in RAW264.7 cells. The Journal of biological chemistry. 2007;282:22834–22847. doi: 10.1074/jbc.M701831200. [DOI] [PubMed] [Google Scholar]
  • 32.Blaho VA, Buczynski MW, Brown CR, Dennis EA. Lipidomic analysis of dynamic eicosanoid responses during the induction and resolution of Lyme arthritis. The Journal of biological chemistry. 2009;284:21599–21612. doi: 10.1074/jbc.M109.003822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Golovko MY, Murphy EJ. An improved LC-MS/MS procedure for brain prostanoid analysis using brain fixation with head-focused microwave irradiation and liquid-liquid extraction. Journal of lipid research. 2008;49:893–902. doi: 10.1194/jlr.D700030-JLR200. [DOI] [PubMed] [Google Scholar]
  • 34.Jian W, Edom R, Weng N, Zannikos P, Zhang Z, Wang H. Validation and application of an LC-MS/MS method for quantitation of three fatty acid ethanolamides as biomarkers for fatty acid hydrolase inhibition in human plasma. Journal of chromatography. 878:1687–1699. doi: 10.1016/j.jchromb.2010.04.024. [DOI] [PubMed] [Google Scholar]
  • 35.Hall LM, Murphy RC. Electrospray mass spectrometric analysis of 5-hydroperoxy and 5-hydroxyeicosatetraenoic acids generated by lipid peroxidation of red blood cell ghost phospholipids. Journal of the American Society for Mass Spectrometry. 1998;9:527–532. doi: 10.1016/S1044-0305(98)00013-0. [DOI] [PubMed] [Google Scholar]

RESOURCES