Abstract
Chronic Post Traumatic Headache (PTH) is among the most common and disabling sequelae of traumatic brain injury (TBI). Current PTH treatments are often only partially effective and have problematic side effects. We previously showed in a small randomized trial of patients with chronic non-traumatic headaches that manipulation of dietary fatty acids decreased headache frequency, severity, and pain medication use. Pain reduction was associated with alterations in oxylipins derived from n-3 and n-6 fatty acids, suggesting that oxylipins could potentially mediate clinical pain reduction. The objective of the present study was to investigate whether circulating oxylipins measured in the acute setting following TBI, could serve as prognostic biomarkers for developing chronic PTH. Participants enrolled in the Traumatic Head Injury Neuroimaging Classification Protocol provided serum within 3 days of TBI and were followed up at 90 days post-injury with a neurobehavioral symptom inventory (NSI) and satisfaction with-life-survey (SWLS). Liquid chromatography tandem mass spectrometry methods profiled 39 oxylipins derived from n-3 docosaheaxaenoic acid (DHA), and n-6 arachidonic acid (AA) and linoleic acid (LA).Statistical analyses assessed the association of oxylipins with headache severity (primary outcome, measured by headache question on NSI) as well as associations between oxylipins and total NSI or SWLS scores. Among oxylipins, 4-hydroxy-DHA and 19,20-epoxy-docosapentaenoate (DHA derivatives) were inversely associated with headache severity, and 11-hydroxy-9-epoxy-octadecenoate (an LA derivative) was positively associated with headache severity. These findings support a potential for DHA-derived oxylipins as prognostic biomarkers for development of chronic PTH.
Introduction
Traumatic Brain Injury (TBI) is among the most common causes of disability in young adults world-wide [10,26,53]. Post-traumatic headache (PTH), a secondary headache disorder that develops within 7 days of a TBI and often resembles a migraine or tension type headache [15], is a common sequelae of TBI. PTH resolves within 90 days in most cases, however about 40% of PTH sufferers continue to have headaches persisting >3 months, thus meeting the criteria for chronic PTH [16,20], Chronic PTH is consistently rated as one of the most consequential and disabling sequelae of TBI, with major adverse impacts on social activities, function and life satisfaction [29,47,51,53,55]. Effective treatments for PTH are lacking, in part due to incomplete understanding of the mechanisms linking acute TBI to chronic PTH [7,11,58]. Identification of prognostic and/or pharmacodynamic biomarkers for PTH could be useful for identifying susceptible subpopulations and targeting/tailoring interventions. Biomarkers associated with PTH can also potentially provide insights into the underlying mechanisms and ultimately, could suggest new targets and, approaches for treatment.
Oxylipins derived from n-3 and n-6 fatty acids as pain mediators
The discovery that non-steroidal anti-inflammatory drugs inhibit the synthesis of prostaglandins from arachidonic acid (AA) [54] identified a specific mechanism linking oxidized lipids (oxylipins) to pain. Advances in mass spectrometry have enabled detection of novel lipids that are present in tissues at nMol concentrations, including several families of oxylipins, derived from omega-6 (n-6) and omega-3 (n-3) polyunsaturated fatty acids (PUFA), that regulate inflammation and pain in preclinical models [14,35,48]. Since humans cannot synthesize double bonds in the n-3 and n-6 position of fatty acids, targeted manipulation of PUFAs by diet or supplementation could potentially alter oxylipins in an analgesic manner. In 2013, we reported, in a small randomized trial of chronic non-traumatic headache patients, that targeted manipulation of dietary n-3 and n-6 PUFA decreased headache frequency and severity, while reducing acute pain medication use [39]. Pain reduction was accompanied by alterations in oxylipins and their precursor n-3 and n-6 PUFA, suggesting that certain oxylipins could potentially mediate clinical pain reduction [38]. These findings were followed up more recently where we demonstrated that circulating concentrations of an oxylipin derived from linoleic acid (LA - a dietary n-6 PUFA) negatively correlated with clinical pain reductions [37]. Additionally, we reported that these oxylipins were elevated in inflamed human tissue, and were involved in nociception in preclinical experiments [37].
Recognizing the growing prevalence of TBI and PTH and the large gaps in the understanding of PTH pathogenesis, we aimed to investigate a potential link between oxylipins and the development of chronic PTH. We hypothesized that oxylipins derived from docosahexaenoic acid (DHA, an n-3 PUFA found in fatty fish), which have been linked to antinociception in preclinical models [30,60,64], would be inversely associated with PTH severity. We measured oxylipins in serum collected during the acute phase following TBI and examined the relationship between oxylipin concentrations and PTH severity 90 days post-TBI. Results from this analysis identified three oxylipins as candidate biomarkers for chronic PTH development.
Methods
Subject Recruitment and Enrollment
This report analyzed a subset of subjects who were enrolled in the Traumatic Head Injury Neuroimaging Classification (“THINC”, NCT01132937). “THINC” is an IRB approved prospective study at 3-trauma centers in the Washington, DC metro area over a 2.5-year period. Patients who presented to the emergency department (ED) following an acute head trauma were enrolled into THINC if they met the following criteria: (1) high clinical suspicion of non-penetrating acute TBI, as determined by the ED physician and for which a head computed tomography (CT) scan was performed, (2) age ≥18 years, (3) interval between time of injury and enrollment <48h, (4) and ability to obtain informed consent from the subject or a legally authorized representative. Data and samples were collected by the investigators of “THINC”-NCT01132937. No research procedures were performed prior to written informed consent. All research subjects included in this analysis agreed to share their data and biospecimens for future research. Authors of this work do not have access to identifiers, and the results of this analysis cannot be linked to specific research participants. Investigators on the collection protocol are aware of this secondary research, have agreed to of the use of the data/sample, but were not scientifically engaged in the collaboration and are, therefore, not listed as authors. We have appropriately acknowledged their contribution.
Blood samples and neurobehavioral symptom inventory (NSI) were collected from eligible patients. Additionally, Glasgow Coma Scale (GCS), potential amnesia, loss of consciousness, relevant history, and clinical diagnoses were entered into standardized case report forms by the THINC team. All patients (or their legally authorized representatives) gave written informed consent for the use of their clinical data and blood samples in accordance with a protocol approved by the Institutional Review Board of the National Institute of Neurological Disorders and Stroke (NINDS). Both blood samples and de-identified demographic and clinical data were provided to the Center for Neuroscience and Regenerative Medicine (CNRM) Biorepository and approval was obtained for use for this project. THINC protocol follow-up included administration of the NSI and the Satisfaction with Life Scale (SWLS). For this report we obtained available serum samples from subjects that had (1) post-injury serum sample collected within 3 days of injury and (2) completed follow-up appointments 90 days post-injury (n=121, Figure 1).
Figure1. Experimental design for the study primary objective and the biosynthetic pathways for oxidized lipid mediators measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS).
(A) Serum samples from patients who suffered from a TBI was analyzed with LC-MS/MS to determine which oxidized lipid mediators were associated with headache severity at 90-days post TBI. (B) the chemical structures of 37 of the 39 oxidized lipid mediators measured by our LC-MS/MS assay (9-hydroxy-octadecatrienoic acid and 18-hydroxy-eicosapentaenoic acid are omitted from the figure). Oxidized lipid mediators are synthesized from n-6 (top panel) or n-3 (bottom panel) PUFA (LA, AA, ALA, EPA, DHA). An arrow connecting lipids symbolizes that one of these lipids can be synthesized from the other either enzymatically or non-enzymatically, with the direction of the reaction indicated by the direction of the arrow. Only oxidized lipids that are measured by our LC-MS/MS assay are depicted with their chemical structures.
TBI, traumatic brain injury; LC-MS/MS, Liquid Chromatography-Tandem Mass Spectrometry; NSI, Neurobehavioral Symptom Inventory; SWLS, Satisfaction with Life Survey; n-6, omega-6; n-3, omega-3; PUFA, polyunsaturated fatty acid; LA, linoleic acid; AA, arachidonic acid; ALA, alpha-linolenic acid; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid; 5-HETE,5-hydroxy-eicosatetraenoic acid; 12-HETE, 12-hydroxy-eicosatetraenoic acid; 15-HETE, 15-hydroxy-eicosatetraenoic acid; 5-oxoETE, 5-keto-eicosatetraenoic acid; LTB4, Leukotriene B4; LXA4, Lipoxin A4; PGE2, Prostaglandin E2; PGF2, Prostaglandin F2; 8-isoPGF2, 8-iso-Prostaglandin F2α; TXB2, Thromboxane B2; 4-HDHA, 4-hydroxy-docosahexaenoic acid; 7-HDHA, 7-hydroxy-docosahexaenoic acid; 10-HDHA,10-hydroxy-docosahexanoic acid; 14-HDHA, 14-hydroxy-docosahexaenoic acid; 17-HDHA, 17-hydroxy-docosahexaenoic acid; 16-EDP, 16-epoxy-docosapentaenoic acid; 19,20-EDP, 19-epoxy-docosapentaenoic acid; MAR1, Maresin1; RVD1, Resolvin D1; PDX, Protectin Dx; PD1, Protectin D1; 9-HODE, 9-hydroxy-octadecenoic acid; 13-HODE, 13-hydroxy-octadecenoic acid; 9-oxoODE, 9-oxo octadecenoic acid; 13-oxoODE, 13-oxo-octadecenoic acid; 9-epOME, 9-epoxy-octadeecnoic acid; 12-epOME, 12-epoxy-octadecenoic acid; 9,10-diHOME, 9,10-dihydroxy-octadecenoic acid; 12,13-diHOME, 12,13-dihydroxy-octadecenoic acid; 9,10,11-triHOME, 9,10,11-trihydroxy-octadecenoic acid; 9,12,13-triHOME, 9,12,13-trihydroxy-octadecenoic acid; 9,10,13-triHOME, 9,10,13-trihydroxy-octadecenoic acid; 9-H-12-E-LA, 9-hydroxy-12-epoxy-octadecenoic acid; 11-H-12-E-LA, 11-hydroxy-12-epoxy-octadecenoic acid; 11-H-9-E-LA, 11-hydroxy-9-epoxy-octadecenoic acid; 13-H-9-E-LA 13-hydroxy-9-epoxy-octadecenoic acid; 9-K-12-E-LA, 9-oxo-12-epoxy octadecenoic acid; 5-HpETE, 5-hydroperoxy-eitosatetraenoic acid; 12-HpETE, 12-hydroperoxy-eitosatetraenoic acid; 15-HpETE, 15-hydroperoxy-eitosatetraenoic acid; 9-HpODE, 9-hydroperoxy-octadecenoic acid; 13-HpODE, 13-hydroperoxy-octadecenoic acid; 4-HpDHA, 4-hydroperoxy-docosahexanoic acid; 7-HpDHA, 7-hydroperoxy-docosahexanoic acid; 14-HpDHA, 14-hydroperoxy-docosahexanoic acid; 17-HpDHA, 17-hydroperoxy-docosahexanoic acid;
Neuroimaging
All subjects obtained non-contrast head CT scans as part of usual clinical care. The clinical CT scans were interpreted by staff radiologists at the participating hospitals, upon hospital admission. Reports were reviewed by study personnel and findings related to trauma were recorded.
TBI Classification
TBI classification was based off of the American Congress of Rehabilitation Medicine definition of mild TBI [19,49]. TBI was classified into 4 categories (silent, mild, moderate, severe) based on a combination of GCS, amount of time the patient was unconscious, amount of post-traumatic amnesia time. A subject was classified as having severe TBI if they met any of the following criteria: 1) GCS between 3 and 8; or 2) >24 hours of both post-traumatic amnesia and >24 hours lost consciousness. If a subject did not meet any criteria for severe TBI they were classified as having a moderate TBI if they presented with either 1) GCS between 9 and 12; 2) >24 hours of post-traumatic amnesia; or 3) 0.5-24 hours of lost consciousness. If a subject did not meet any criteria for a moderate TBI they were assessed as having a mild TBI if they presented with either 1) GCS of 13-15; 2) <24 hours of post-traumatic amnesia; or 3) less than 30 mins of lost consciousness. A patient was classified as having a silent TBI if they did not meet any criteria for severe, moderate or mild TBI.
Blood Sampling
Average time from injury to blood collection was 16 hours and all samples were collected within 61.5 hours after injury. Blood was collected from an 18G antecubital vein catheter into serum separator tubes and stored at 4 °C until serum isolation.
Serum Processing
The serum processing, solid phase extraction (SPE) and LC-MS/MS methods have been described in detail [37,52,63]. Briefly, patient serum (200 μL) was thawed and aliquoted into 500 μL ice cold methanol containing 0.02% butylated hydroxy toluene (BHT) and 0.02% ethylenediaminetetraacetic acid (EDTA) and 10 ng/mL of deuterated internal standards. Samples were then incubated at −80°C for 1 hour, to precipitate proteins, and then centrifuged at 17000 x g for 10 min at 4°C to create a protein pellet. The supernatant was collected and stored under N2 gas at −80°C overnight.
Solid Phase Extraction
Methanol extracts of serum samples were diluted in 6 mL of ice cold, ultrapure water and loaded onto SPE columns (Strata™-X 33 μm Polymeric Reversed Phase, 200 mg, Phenomenex, CA) that had been conditioned with methanol, followed by water. Once the samples were loaded onto SPE columns, the columns were cleaned with 10% methanol and allowed to dry for 2 min. Oxidized lipids were eluted, with methanol containing 0.0004% BHT, into glass culture tubes containing 10 μL of 30% glycerol in methanol. Samples were then concentrated by completely evaporating the methanol under N2 gas and reconstituting into 40 μL HPLC grade methanol. The whole sample was then transferred into a 250 μL glass insert that was fit into a 2 mL amber vial (Agilent, CA) for LC-MS/MS analysis.
Liquid Chromatography Tandem-Mass Spectrometry (LC-MS/MS)
Detailed descriptions of the LC-MS/MS conditions and parameters have previously been described by our group [63]. Data processing was performed with Analyst Software™ (version 1.6.3), with the limit of quantitation (LOQ) for each analyte assessed by manual peak inspection in explorer mode of the Analyst Software™. The LOQ was defined as the lowest point of the standard curve, or lowest sample with a signal to noise ratio (S/N) of greater than 5. The LOQ was first established by using the Analyst Software calculated concentration of the sample (i.e. finding the lowest concentration sample with S/N >5). Next samples with analyte peak areas lower than that of the sample defining the LOQ were manually inspected to confirm a S/N ≥5. For analytes with an 18-carbon backbone, samples with calculated concentrations or peak areas lower than the lowest standard within the linear range of our calibration curve were classified as below the limit of quantitation (BLQ) and the LOQ was defined as the concentration of the lowest standard curve sample within the linear range. Calibration curves tended to be linear below the LOQ for analytes with a backbone greater than 18 carbons. Samples determined to be BLQ were imputed to have a concentration of ½ LOQ. After chromatogram inspection we determined that the conditions of our LC-MS/MS were unstable during 12 sample runs (an approximately 12-hour window that occurred sequentially): these samples exhibited large shifts in peak retention times, that were not predictable by the shifting of the internal standard peaks. Due to these unpredictable shifting retention times we were not able to identify analytes of interest with confidence. Therefore, samples that were run during the abnormal LC-MS/MS conditions were classified as missing completely at random and excluded from the analysis. This designation—missing completely at random—indicates a situation in which the missing values are unlikely to be related to outcome variables [24].
Neurobehavioral Symptom Inventory and Satisfaction with Life Scale
The primary outcome was headache severity which was extracted from the NSI. The NSI is a 22-item questionnaire used by clinicians to quantify TBI symptom severity [5]. Each NSI question refers to a TBI related symptom (e.g., “Headaches”) and asks the patient to rate how that symptom has affected their life on a 0-4 scale (0 = “Rarely if ever present; not a problem at all” and 4 = “Almost always present and I have been unable to perform at work, school or home due to this”). Our primary endpoint and our exploratory endpoints were headache severity (headache question of the NSI) and the total NSI score, respectively, with both assessed at 90 days post TBI. The headache question of the NSI has been reported to be well correlated with the Headache Impact Test (HIT-6) [23]. We also explored the relationship between oxylipins following TBI and life satisfaction at 90-days post TBI using the SWLS. SWLS is a 5-item questionnaire that is used to assess global life satisfaction [8]. Each item on the SWLS was rated by the participant on a scale of 1 (strongly disagree) to 7 (strongly agree) with a total score between 26 and 30 indicating the participant was satisfied with their life. The SWLS has been shown to be appropriate for a wide range of applications [33], including evaluation of quality-of-life in TBI populations [2].
Using archival samples to investigate whether candidate biomarkers are pharmacodynamic
To examine the potential for targeted dietary (n-3 and n-6 PUFA) manipulations to modify candidate biomarkers identified in this report, we measured concentrations of oxylipins, using the above described methods, in archived baseline and post-intervention plasma samples (n=44) collected from the chronic daily headache trial (NCT01157208). The methods (including CONSORT diagram) and results of this small randomized trial have been published elsewhere [38-40]. Briefly, a high n-3 plus low n-6 dietary PUFA intervention decreased headache frequency and severity, while increasing precursor n-3 and n-6 fatty acids and selected oxylipins. Here we quantified an expanded list of n-3 and n-6 PUFA derived oxylipins to investigate whether circulating levels of these candidate biomarkers are modifiable by diet and to explore whether diet-induced changes correlate with clinical pain reduction.
Statistics
We excluded 12 subjects with abnormal MS as missing completely at random. Missing oxylipin values were imputed with ½ of the limit of quantitation. We investigated the effect of each serum post-injury oxylipin on each outcome at day 90 using three types of models: ordinal regression for NSI headache severity, negative binomial regression for NSI total score, and linear regression for the total SWLS. Each model was adjusted for age, gender, race, ethnicity, observed CT scan abnormalities that were consistent with TBI, THINC TBI rating (silent, mild, moderate, or severe), and time from injury to blood draw as well as the log sum of oxylipins from each precursor fatty acid (LA, DHA, and ARA) excluding the oxylipin in question. Model variables were chosen a priori. Analyses were performed in Stata 15 (StataCorp. 2017. Stata Statistical Software: Release 15. College Station, TX: StataCorp LLC.).
Results
Population Characteristics
Demographics and clinical characteristics for our TBI study population are shown in Table 1. Patients enrolled in this study were 69% male and 74% White. Most of the injuries were classified as a mild TBI (71%), with only a single incidence of severe TBI. Demographics and clinical characteristics of the chronic daily headache cohort have been previously published [39].
Table 1.
Population Demographics and Clinical Characteristics (n=121)
| Characteristic | Mean or Count |
SD or % |
|---|---|---|
| Years of Age | 48 | 18 |
| Male | 84 | 69% |
| Race | ||
| Black | 27 | 22% |
| White | 89 | 74% |
| Asian | 4 | 3% |
| Native American | 1 | 1% |
| Latino or Hispanic | 19 | 16% |
| Hours from Injury to Blood Draw | 16 | 12 |
| TBI Found on CT Scan | 38 | 31% |
| THINC TBI Rating | ||
| Silent | 26 | 21% |
| Mild | 86 | 71% |
| Moderate | 8 | 7% |
| Severe | 1 | 1% |
SD, standard deviation; TBI, Traumatic Brain Injury; CT, Computed Tomography; THINC, Traumatic Head Injury Neuroimaging Classification
Post-Traumatic Headache severity decreases with time
Median headache severity declined over time (Figure 2a). In the acute phase following TBI, 78% of participants reported having any headache, while only 40% did at 90 days post-injury.
Figure 2. Three oxylipins are associated with chronic headache severity.
(A) Immediately following TBI, median headache severity was 3 out of 4 and it declined over time. (B) Three oxylipins were significantly associated with headache severity at 90 days post-TBI. There was a higher probability of having no headaches at 90 days with higher serum concentrations of (C) 4-HDHA and (D) 19,20-EDP (p=0.005 and p=0.019, respectively) and lower concentrations of (E) 11-H-9-E-LA (p=0.031). Standardized coefficients (B) and probabilities (C) are based on ordinal logistic regression of day 90 NSI headache question on each respective serum post-injury mediator, adjusted for the sums of LA, DHA, and AA mediators, age, gender, race, ethnicity, traumatic brain injury, and time from injury to blood draw. This analysis excludes 12 subjects with abnormal mass spectrometry (n=109).
IQR, interquartile range; NSI, neurobehavioral symptom inventory; TBI, Traumatic Brain Injury; V. Severe, very severe; 4-HDHA, 4-hydroxy-docosahexaenoic acid; 19,20-EDP, 19-epoxy-docosapentaenoic acid; 11-H-9-E-LA, 11-hydroxy-9-epoxy-octadecenoic acid; ALA, alpha-linolenic acid; LA, linoleic acid; EPA, eicosapentaenoic acid; DHA, docosahexaenoic acid; AA, arachidonic acid;
Concentrations of oxidized lipid mediators in serum
We profiled for 39 oxylipins, 27 of which were detected in a sufficient number of samples to include in our analysis (Figure 1). These include several classic AA-derived lipid mediators of pain and inflammation (i.e., PGE2, TXB2, LTB4), two DHA-derived oxylipins (14-HDHA and 17-HDHA) that are pathway precursors for specialized pro-resolving lipid mediators with anti-nociceptive properties in preclinical models, and several LA-derived oxylipins that are reported to have pro-nociceptive properties in preclinical models.
Serum oxylipin concentrations are listed in Supplementary Table 1. In general, oxylipins derived from n-6 PUFA (AA or LA) were more highly concentrated than n-3 PUFA derived oxylipins (Supplementary Table 1). Of the classic mediators of pain and inflammation that we were able to measure, TXB2 was approximately 30 and 60-fold more concentrated in serum compared to LTB4 and PGE2, respectively (Supplementary Table 1). Notably, 8-iso Prostaglandin F2α (8-IsoPGF2a), a marker of non-enzymatic peroxidation, was not detected in any sample.
Oxylipins associated with headache severity following TBI
Only three of the oxylipins measured were significantly associated with headache severity at 90 days post-TBI (Figure 2b). Two DHA-derived oxylipins, 4-HDHA and 19,20-EDP (Figure 2c and d), were inversely associated with headache severity (p≤0.019). One LA-derived oxylipin, 11-H-9,10-E-LA, was positively associated with headache severity (p<0.031 Figure 2e). To put this in context of the serum concentrations observed, a one standard deviation increase in 4-HDHA and 19,20-EDP was associated with a 70 and 60% decrease, respectively, in the odds of having headache at 90 days, while a one standard deviation increase in 11-H-9,10-E-LA was associated with a 90% increase.
Oxylipins associated with overall traumatic brain injury symptoms and life satisfaction
Associations between serum oxylipins after TBI and total NSI score, which measures TBI symptom severity, 90 days after TBI were weak (figure 3). 13-HODE (p=0.037) and 4-HDHA (p=0.048) were the only oxylipins that were associated with NSI score at a p<0.05 level [β (95% CI) = −0.38 (−0.73, −0.02) and −0.33 (−0.66, −0.00), respectively).
Figure 3. Association of post-injury oxylipins and NSI total score at day 90 (n=109).

Acute serum concentration of 1 oxylipin (13-HODE) was inversely associated with total NSI. Incident ratios were estimated using negative binomial regression of the day-90 NSI score on each respective serum post-injury oxylipin, adjusted for age, gender, race, ethnicity, traumatic brain injury (THINC and CT scan) and time from injury to blood draw. 12 subjects with abnormal MS were excluded. Coefficients are adjusted for log sums of LA, DHA and ARA derived oxylipin concentrations, excluding the mediator in question. 5-HETE,5-hydroxy-eicosatetraenoic acid; 12-HETE, 12-hydroxy-eicosatetraenoic acid; 15-HETE, 15-hydroxy-eicosatetraenoic acid; 5-oxoETE, 5-keto-eicosatetraenoic acid; LTB4, Leukotriene B4; PGE2, Prostaglandin E2; TXB2, Thromboxane B2; 4-HDHA, 4-hydroxy-docosahexaenoic acid; 10-HDHA,10-hydroxy-docosahexanoic acid; 14-HDHA, 14-hydroxy-docosahexaenoic acid; 17-HDHA, 17-hydroxy-docosahexaenoic acid; 19,20-EDP, 19,20-epoxy-docosapentaenoic acid; 9-HODE, 9-hydroxy-octadecenoic acid; 13-HODE, 13-hydroxy-octadecenoic acid; 13-oxoODE, 13-oxo-octadecenoic acid; 9,10-epOME, 9-epoxy-octadeecnoic acid; 12,13-epOME, 12-epoxy-octadecenoic acid; 9,10-diHOME, 9,10-dihydroxy-octadecenoic acid; 12,13-diHOME, 12,13-dihydroxy-octadecenoic acid; 9,10,11-triHOME, 9,10,11-trihydroxy-octadecenoic acid; 9,12,13-triHOME, 9,12,13-trihydroxy-octadecenoic acid; 9,10,13-triHOME, 9,10,13-trihydroxy-octadecenoic acid; 11-H-12-E-LA, 11-hydroxy-12-epoxy-octadecenoic acid; 11-H-9-E-LA, 11-hydroxy-9-epoxy-octadecenoic acid; 13-H-9-E-LA, 13-hydroxy-9-epoxy-octadecenoic acid.
In another exploratory analyses, post-injury 12,13 DiHOME and 14-HDHA were associated [β (95% CI) = −0.39 (−0.67, −0.11) and 0.36 (0.06, 0.65), respectively) with SWLS (figure 4) at 90 days post-injury (p=0.007 and 0.018, respectively).
Figure 4. Association of post-injury lipid mediators and satisfaction with life score (SWLS) at day 90 (n=109).

Standardized coefficients were estimated using negative linear regression of the day-90 SWLS score on each respective serum post-injury oxylipin, adjusted for age, gender, race, ethnicity, traumatic brain injury (THINC and CT scan) and time from injury to blood draw. 12 subjects with abnormal MS were excluded. Coefficients are adjusted for log sums of LA, DHA and ARA derived oxylipin concentrations, excluding the mediator in question. 5-HETE,5-hydroxy-eicosatetraenoic acid; 12-HETE, 12-hydroxy-eicosatetraenoic acid; 15-HETE, 15-hydroxy-eicosatetraenoic acid; 5-oxoETE, 5-keto-eicosatetraenoic acid; LTB4, Leukotriene B4; PGE2, Prostaglandin E2; TXB2, Thromboxane B2; 4-HDHA, 4-hydroxy-docosahexaenoic acid; 10-HDHA,10-hydroxy-docosahexanoic acid; 14-HDHA, 14-hydroxy-docosahexaenoic acid; 17-HDHA, 17-hydroxy-docosahexaenoic acid; 19,20-EDP, 19,20-epoxy-docosapentaenoic acid; 9-HODE, 9-hydroxy-octadecenoic acid; 13-HODE, 13-hydroxy-octadecenoic acid; 13-oxoODE, 13-oxo-octadecenoic acid; 9,10-epOME, 9-epoxy-octadeecnoic acid; 12,13-epOME, 12-epoxy-octadecenoic acid; 9,10-diHOME, 9,10-dihydroxy-octadecenoic acid; 12,13-diHOME, 12,13-dihydroxy-octadecenoic acid; 9,10,11-triHOME, 9,10,11-trihydroxy-octadecenoic acid; 9,12,13-triHOME, 9,12,13-trihydroxy-octadecenoic acid; 9,10,13-triHOME, 9,10,13-trihydroxy-octadecenoic acid; 11-H-12-E-LA, 11-hydroxy-12-epoxy-octadecenoic acid; 11-H-9-E-LA, 11-hydroxy-9-epoxy-octadecenoic acid; 13-H-9-E-LA, 13-hydroxy-9-epoxy-octadecenoic acid.
Analysis of archived serum samples from 2013 dietary headache intervention trial
The primary results of the chronic daily headache diet trial dietary headache were published in 2013 [38,39]. Briefly, a 12-week intervention altering n-3 and n-6 PUFA led to clinically meaningful reductions in headache impact and severity [39]. Here we found that these interventions significantly increased plasma concentrations of 4-HDHA (p=0.03) and 19,20-EDP (p=0.002) (figure 5). Additionally, diet-induced changes in both 4-HDHA and 19,20-EDP were inversely associated with headache hours per day, headache days per month and headache impact test (HIT-6) (figure 5, p<0.007). The results of this analysis for 11-H-9,10-E-LA have been published elsewhere [37]. Briefly, diet did not significantly alter circulating 11-H-9,10-E-LA and this oxylipin was not associated with any headache outcome measured [37]. With respect to other oxylipins measured in this analysis, we observed that DHA derived oxylipins were inversely associated with headache impact and severity while only 17-HDHA was appreciably altered by diet (Supplementary Table 2). Notably, 8-isoPGF2a was detected in only 5 samples and median 4-HDHA and 19,20-EDP concentrations were similar to those of our PTH cohort (0.04 and 0.05 ng/ml, respectively) indicating substantial lipid peroxidation was not occurring in these samples.
Figure 5. Analysis of archival serum samples from 2013 dietary headache trial indicate that 4-hydroxy docosahexanoic acid (4-HDHA) and 19,20-epoxy docosapentaenoic acid (19,20-EDP) are modifiable by diet and support their relation to headache severity.
(A) Serum concentrations of 4-HDHA and 19,20-EDP were increased (p=0.03 and 0.002, respectively) after 12 weeks where participants were fed a low omega-6 PUFA diet, with or without increased omega-3 PUFA. After 12-weeks on their respective diet, participants (B) headache hours per day, (C) headache days per month and (D) Headache Impact Test (HIT-6) scores were inversely associated with serum oxylipin concentrations (p<0.007).
Discussion
Summary and context:
Chronic PTH is among the most common and disabling sequelae of TBI. Mechanisms underlying the transition from acute TBI to chronic PTH are poorly understood and prognostic, mechanism-based and pharmacodynamic biomarkers for chronic PTH are lacking. Since oxylipins derived from n-3 and n-6 PUFA play key roles in the amplification and resolution of inflammatory responses and nociception [14,48] and can be manipulated by diet [41], circulating oxylipins are intriguing candidates for non-invasive, mechanism-based, pharmacodynamic biomarkers with predictive power for the development and resolution of chronic pain. In the present study, we identified two oxylipins derived from DHA (4-HDHA and 19,20-EDP) measured in serum during the acute phase following TBI that were strongly, inversely associated with risk of developing chronic PTH, and one LA derived oxidized lipid mediator (11-H-9-E-LA) that was positively, but relatively weakly, associated with headache severity. These oxylipins can potentially serve as biomarkers in a clinical setting to identify TBI patients that may develop chronic PTH.
Mechanism-based biomarker candidates
The main objective of this work was to identify biomarker candidates for chronic PTH following TBI. However, plausible biological mechanisms could potentially help explain these observed associations between oxidized derivatives of DHA and LA and chronic PTH. In preclinical models, oxidized derivatives of DHA—including DHA-derived mono-epoxides [57], resolvins [30,60], protectins [46], and maresins [3,64]—are reported to have potent anti-inflammatory and anti-nociceptive effects. By contrast, in preclinical models oxidized derivatives of LA—including HODEs, DiHOMEs, and hydroxy-epoxides—are reported to have pro-nociceptive properties [12,31,32,37,65]. The exact cause of chronic PTH is unknown, however, the findings reported here indicate that the relative levels of n-3 and n-6 PUFA derived oxylipins, immediately after injury, may play a role in disease development. Future work should investigate the biological effects of oxylipins in TBI.
Pharmacodynamic biomarkers (responsive to diet)
We previously showed in a small randomized trial of patients with chronic non-traumatic headaches that targeted manipulation of dietary PUFA—increasing n-3 EPA + DHA while decreasing LA—decreased headache frequency and severity [39]. Here, using archived samples from that small trial, we expand upon those findings by demonstrating that 4-HDHA and 19,20-EDP are pharmacodynamic (i.e. altered by the targeted substrate manipulation) and that diet-induced changes in these two oxylipins were closely correlated with clinical pain reduction. These findings provide further support for these two compounds as candidate prognostic biomarkers of pain reduction. In an RCT in 60 chronic migraine sufferers, coadministration of an EPA and DHA supplement with amitriptyline (a tricyclic antidepressant with analgesic properties) decreased the number of headache days per month compared to placebo and amitriptyline [50]. However, in a larger RCT in patients with episodic migraines, fish oil supplementation provided no benefit compared to placebo [36]. Thus, collective findings from randomized trials testing dietary or supplemental provision of EPA + DHA on headache outcomes are mixed and may differ depending on accompanying pharmacological interventions or dietary changes. Two larger RCTs testing the effects of targeted substrate manipulation—increasing EPA+DHA with or without LA lowering—on pain and related endpoints in patients with frequent episodic migraines (ClinicalTrials.gov Identifier: NCT02012790 [25]) and chronic (unresolved) PTH (ClinicalTrials.gov Identifier: NCT03272399) will help clarify the biochemical and clinical consequences of these interventions and the potential utility of these oxylipins as pharmacodynamic, mechanism-based biomarkers for these conditions.
Effects of TBI on lipid metabolism
It has been reported that lipid metabolism is impacted by TBI [1,44]. In rodents, after a TBI cortical concentrations of esterified pools of DHA have been reported to decrease acutely and then gradually increase in a manner suggestive of a role of DHA containing phospholipids in injury healing and recovery [13]. Unesterified DHA is believed to be the form of DHA that most abundantly enters the brain [4] and in humans, acutely after an injury this form of DHA is increased in the plasma and CSF [9,34] suggesting an increased uptake of DHA into the brain. Interestingly, studies in mice have found that increasing plasma DHA concentrations after an injury is associated with improved TBI recovery [22] suggesting a role for DHA in treating TBI. Despite this preclinical evidence, use of n-3 PUFA to treat TBI remains mostly anecdotal [21]. Our findings that DHA derived oxylipins were associated with less headache severity and greater life satisfaction in humans support this hypothesis, however, we cannot speculate on the cause of elevated oxidized DHA derived lipids in patients with better outcomes (i.e. diet/supplement vs. genetics).
TBI has been shown to alter lipid peroxidation and oxidized lipid concentrations in blood and CSF [9,17,59,61,62]. Previous reports in humans have indicated that in CSF oxylipins derived from AA tend to be increased after TBI [9,56,59]. Results from our study do not provide support for the use of circulating AA derived oxylipins (i.e. prostaglandins, leukotrienes and mono-oxygenated AA derivatives) as prognostic biomarkers of TBI recovery. It should be noted, however, that most reports linking AA-derived oxylipins to TBI measured oxylipins in CSF of TBI cases vs controls making it impossible to compare to this report, where we analyzed oxylipins solely in serum of TBI patients.
To our knowledge this is the first report to profile LA-derived oxylipins in TBI using LC-MS/MS. A recent report found that serum LA was lower in TBI patients with cognitive impairment compared to those without cognitive impairment [61] but did not measure LA-derived oxylipins. In the present study, higher concentrations of the LA-derived oxylipins 11-H-9-E-LA and 12,13-DiHOME were associated with chronic PTH severity and lower life satisfaction recovery, respectively. However, higher concentrations of another LA-derived oxylipin (13-HODE) were associated with improved TBI recovery. Given the number of statistical comparisons, it is possible that these discordant associations are simply due to chance. Future studies are needed to better understand the potential roles of LA-derived oxylipins in TBI and PTH.
It should be stressed that oxylipins were identified as potential biomarkers of chronic PTH development, and the observational design of our study did not allow us to draw conclusions on their bioactivity. Particularly, we were unable to conclude if these oxylipins were related specifically to brain injury or general tissue injury, since TBI is often accompanied by damage to other tissues [6]. Traumatic (non-brain specific) injury can increase synthesis of oxylipins [18,43], and expression of genes encoding enzymes that catalyze oxylipin synthesis were associated with complications from traumatic injury [28]. We did not collect information relating to extent of non-brain injuries in our participants nor did we analyze serum of non-TBI patients, therefore, we are unable to conclude on the relationship of TBI with oxylipin concentrations. Additionally, oxylipins were measured acutely after TBI and not at the time of our clinical endpoint (3 months post-TBI), therefore, future study is required to draw conclusions about the bioactivity or mechanism of action of these oxylipins in TBI and headache pathophysiology. Nevertheless, we identified 2 oxylipins (4-HDHA and 19,20-EDP) acutely after TBI that were associated with the development of chronic headache. Identifying signatures that predict the transition of acute to chronic pain is of great importance clinically and scientifically, and the need for this research is reflected in the fact that an NIH Common Fund Program (The Acute to Chronic Pain Signatures Program) is dedicated to this research [27].
Limitations
Several limitations of the present work require consideration. Since this is an observational study, associations reported here should not be interpreted as causal. Our measure for headache severity was extracted from a larger questionnaire (NSI) designed to evaluate total overall brain injury symptoms and not specifically headache. However, the NSI headache question has been reported to be significantly correlated with HIT-6 (r=0.8) and to adequately capture headache severity [23]. Future work assessing this hypothesis should utilize a questionnaire specifically designed and validated to assess headache severity. Additionally, only 8% of the TBI cases in this report were rated moderate or severe. Though the majority of TBI cases are of mild severity [10], future work should aim to select for moderate and severe TBI cases. Samples were collected in a clinical setting; thus it is possible that delays between sample collection and processing to serum could have impacted oxylipin results. Previously published reports indicate that oxylipins are stable in blood stored at 4°C for up to 4 hours [42,45]. Though we were unable to record blood processing times, oxylipins generated via non-enzymatic lipid peroxidation (i.e. 8-isoPGF2a) were BLQ (LOQ = 0.1ng/ml) in all PTH cohort sample indicating that substantial auto-oxidation likely did not occur. Additionally, median oxylipin concentrations in our PTH cohort and in our archival samples were remarkably consistent with a previous report from our group that measured oxylipins in a research setting [52] indicating that blood processing or storage did not significantly impact oxylipin concentrations. Limitations that are common to mass spectrometry assays—including lack of resolution of isomers and related compounds, could potentially influence results. Finally, these results require confirmation in larger studies.
In summary, we identified two DHA-derived oxylipins as mechanism-based, pharmacodynamic biomarker candidates for predicting the development of chronic PTH following TBI. Future studies are needed to validate these findings.
Supplementary Material
Acknowledgements
We wish to acknowledge the staff at Suburban Hospital and Washington Hospital Center, and the Investigators of the Center for Neuroscience and Regenerative Medicine THINC Study team and particpants. Support for this work included funding from Department of Defense in the Center for Neuroscience and Regenerative Medicine and the intramural programs of the National Institute on Aging and National Institute on Alcohol Abuse and Alcoholism.
Footnotes
Conflicts of Interest: None
Disclosures: Partial and preliminary results of this manuscript were presented at the 13th Congress of the International Society for the Study of Fatty Acids and Lipids as well as at the 2019 National Capital Area TBI Research Symposium.
Authors have no conflicts of interest to declare
References
- [1].Anthonymuthu TS, Kenny EM, Amoscato AA, Lewis J, Kochanek PM, Kagan VE, Bayır H. Global assessment of oxidized free fatty acids in brain reveals an enzymatic predominance to oxidative signaling after trauma. Biochim. Biophys. Acta Mol. Basis Dis 2017;1863:2601–2613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Braden CA, Cuthbert JP, Brenner L, Hawley L, Morey C, Newman J, Staniszewski K, Harrison-Felix C. Health and wellness characteristics of persons with traumatic brain injury. Brain Inj. 2012;26:1315–1327. [DOI] [PubMed] [Google Scholar]
- [3].Cezar TLC, Martinez RM, da Rocha C, Melo CPB, Vale DL, Borghi SM, Fattori V, Vignoli JA, Camilios-Neto D, Baracat MM, Georgetti SR, Verri WA, Casagrande R. Treatment with maresin 1, a docosahexaenoic acid-derived pro-resolution lipid, protects skin from inflammation and oxidative stress caused by UVB irradiation. Sci. Rep 2019;9:3062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Chen CT, Kitson AP, Hopperton KE, Domenichiello AF, Trépanier M-O, Lin LE, Ermini L, Post M, Thies F, Bazinet RP. Plasma non-esterified docosahexaenoic acid is the major pool supplying the brain. Sci. Rep 2015;5:15791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Cicerone KD, Kalmar K. Persistent postconcussion syndrome: The structure of subjective complaints after mild traumatic brain injury. J. Head Trauma Rehabil 1995;10:1–17. [Google Scholar]
- [6].Dacey R, Dikmen S, Temkin N, McLean A, Armsden G, Winn HR. Relative effects of brain and non-brain injuries on neuropsychological and psychosocial outcome. J. Trauma 1991;31:217–222. [PubMed] [Google Scholar]
- [7].Defrin R. Chronic post-traumatic headache: clinical findings and possible mechanisms. J Man Manip Ther 2014;22:36–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Diener E, Emmons RA, Larsen RJ, Griffin S. The Satisfaction With Life Scale. J. Pers. Assess 1985;49:71–75. [DOI] [PubMed] [Google Scholar]
- [9].Farias SE, Heidenreich KA, Wohlauer MV, Murphy RC, Moore EE. Lipid mediators in cerebral spinal fluid of traumatic brain injured patients. J. Trauma 2011;71:1211–1218. [DOI] [PubMed] [Google Scholar]
- [10].Feigin VL, Theadom A, Barker-Collo S, Starkey NJ, McPherson K, Kahan M, Dowell A, Brown P, Parag V, Kydd R, Jones K, Jones A, Ameratunga S, BIONIC Study Group. Incidence of traumatic brain injury in New Zealand: a population-based study. Lancet Neurol. 2013;12:53–64. [DOI] [PubMed] [Google Scholar]
- [11].Formisano R, Bivona U, Catani S, D’Ippolito M, Buzzi MG. Post-traumatic headache: facts and doubts. J Headache Pain 2009;10:145–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Green D, Ruparel S, Gao X, Ruparel N, Patil M, Akopian A, Hargreaves K. Central activation of TRPV1 and TRPA1 by novel endogenous agonists contributes to mechanical allodynia and thermal hyperalgesia after burn injury. Mol. Pain 2016;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Guo S, Zhou D, Zhang M, Li T, Liu Y, Xu Y, Chen T, Li Z. Monitoring changes of docosahexaenoic acid-containing lipids during the recovery process of traumatic brain injury in rat using mass spectrometry imaging. Sci. Rep 2017;7:5054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Hargreaves KM, Ruparel S. Role of oxidized lipids and TRP channels in orofacial pain and inflammation. J. Dent. Res 2016;95:1117–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Headache Classification Committee of the International Headache Society (IHS). The International Classification of Headache Disorders, 3rd edition. Cephalalgia 2018;38:1–211. [DOI] [PubMed] [Google Scholar]
- [16].Hoffman JM, Lucas S, Dikmen S, Braden CA, Brown AW, Brunner R, Diaz-Arrastia R, Walker WC, Watanabe TK, Bell KR. Natural history of headache after traumatic brain injury. J. Neurotrauma 2011;28:1719–1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Kasprzak HA, Woźniak A, Drewa G, Woźniak B. Enhanced lipid peroxidation processes in patients after brain contusion. J. Neurotrauma 2001;18:793–797. [DOI] [PubMed] [Google Scholar]
- [18].Kaufmann I, Hauer D, Huge V, Vogeser M, Campolongo P, Chouker A, Thiel M, Schelling G. Enhanced anandamide plasma levels in patients with complex regional pain syndrome following traumatic injury: a preliminary report. Eur Surg Res 2009;43:325–329. [DOI] [PubMed] [Google Scholar]
- [19].Kay T, Harrington DE, Adams R, Anderson T, Berrol S, Cicerone K, Dahlberg C, Gerber D, Goka R, Harley P, Hilt J, Horn L, Lehmkuhl D, Malec J. Definition of mild traumatic brain injury. J Head Trauma Rehabil 1993;8:86–87. [Google Scholar]
- [20].Lahz S, Bryant RA. Incidence of chronic pain following traumatic brain injury. Arch. Phys. Med. Rehabil 1996;77:889–891. [DOI] [PubMed] [Google Scholar]
- [21].Lewis M, Ghassemi P, Hibbeln J. Therapeutic use of omega-3 fatty acids in severe head trauma. Am. J. Emerg. Med 2013;31:273.e5–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].López-Vales R, Redensek A, Skinner TAA, Rathore KI, Ghasemlou N, Wojewodka G, DeSanctis J, Radzioch D, David S. Fenretinide promotes functional recovery and tissue protection after spinal cord contusion injury in mice. J. Neurosci 2010;30:3220–3226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Lu LH, Bowles AO, Kennedy JE, Eapen BC, Cooper DB. Single-Item Versus Multiple-Item Headache Ratings in Service Members Seeking Treatment for Brain Injury. Mil. Med 2020;185:e43–e46. [DOI] [PubMed] [Google Scholar]
- [24].Mallinckrodt C. Preventing and treating missing data in longitudinal clinical trials: A practical guide. Cambridge: Cambridge University Press, 2013. [Google Scholar]
- [25].Mann JD, Faurot KR, MacIntosh B, Palsson OS, Suchindran CM, Gaylord SA, Lynch C, Johnston A, Maiden K, Barrow DA, Hibbeln JR, Ramsden CE. A sixteen-week three-armed, randomized, controlled trial investigating clinical and biochemical effects of targeted alterations in dietary linoleic acid and n-3 EPA+DHA in adults with episodic migraine: Study protocol. Prostaglandins Leukot Essent Fatty Acids 2018;128:41–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Moye LS, Pradhan AA. From blast to bench: A translational mini-review of posttraumatic headache. J. Neurosci. Res 2017;95:1347–1354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].National Institutes of Health. Acute to Chronic Pain Signatures ∣ NIH Common Fund. Acute to Chronic Pain Signatures (A2CPS) 2020. Available: https://commonfund.nih.gov/pain. Accessed 5 May 2020.
- [28].Orr SK, Butler KL, Hayden D, Tompkins RG, Serhan CN, Irimia D. Gene expression of proresolving lipid mediator pathways is associated with clinical outcomes in trauma patients. Crit. Care Med 2015;43:2642–2650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Packard RC. Chronic post-traumatic headache: associations with mild traumatic brain injury, concussion, and post-concussive disorder. Curr Pain Headache Rep 2008;12:67–73. [DOI] [PubMed] [Google Scholar]
- [30].Park C-K, Xu Z-Z, Liu T, Lü N, Serhan CN, Ji R-R. Resolvin D2 is a potent endogenous inhibitor for transient receptor potential subtype V1/A1, inflammatory pain, and spinal cord synaptic plasticity in mice: distinct roles of resolvin D1, D2, and E1. J. Neurosci 2011;31:18433–18438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Patwardhan AM, Akopian AN, Ruparel NB, Diogenes A, Weintraub ST, Uhlson C, Murphy RC, Hargreaves KM. Heat generates oxidized linoleic acid metabolites that activate TRPV1 and produce pain in rodents. J. Clin. Invest 2010;120:1617–1626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Patwardhan AM, Scotland PE, Akopian AN, Hargreaves KM. Activation of TRPV1 in the spinal cord by oxidized linoleic acid metabolites contributes to inflammatory hyperalgesia. Proc. Natl. Acad. Sci. USA 2009;106:18820–18824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Pavot W, Diener E, Colvin CR, Sandvik E. Further validation of the Satisfaction with Life Scale: evidence for the cross-method convergence of well-being measures. J. Pers. Assess 1991;57:149–161. [DOI] [PubMed] [Google Scholar]
- [34].Pilitsis JG, Coplin WM, O’Regan MH, Wellwood JM, Diaz FG, Fairfax MR, Michael DB, Phillis JW. Free fatty acids in cerebrospinal fluids from patients with traumatic brain injury. Neurosci. Lett 2003;349:136–138. [DOI] [PubMed] [Google Scholar]
- [35].Piomelli D, Hohmann AG, Seybold V, Hammock BD. A lipid gate for the peripheral control of pain. J. Neurosci 2014;34:15184–15191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Pradalier A, Bakouche P, Baudesson G, Delage A, Cornaille-Lafage G, Launay JM, Biason P. Failure of omega-3 polyunsaturated fatty acids in prevention of migraine: a double-blind study versus placebo. Cephalalgia 2001;21:818–822. [DOI] [PubMed] [Google Scholar]
- [37].Ramsden CE, Domenichiello AF, Yuan Z-X, Sapio MR, Keyes GS, Mishra SK, Gross JR, Majchrzak-Hong S, Zamora D, Horowitz MS, Davis JM, Sorokin AV, Dey A, LaPaglia DM, Wheeler JJ, Vasko MR, Mehta NN, Mannes AJ, Iadarola MJ. A systems approach for discovering linoleic acid derivatives that potentially mediate pain and itch. Sci. Signal 2017;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Ramsden CE, Faurot KR, Zamora D, Palsson OS, MacIntosh BA, Gaylord S, Taha AY, Rapoport SI, Hibbeln JR, Davis JM, Mann JD. Targeted alterations in dietary n-3 and n-6 fatty acids improve life functioning and reduce psychological distress among patients with chronic headache: a secondary analysis of a randomized trial. Pain 2015;156:587–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Ramsden CE, Faurot KR, Zamora D, Suchindran CM, Macintosh BA, Gaylord S, Ringel A, Hibbeln JR, Feldstein AE, Mori TA, Barden A, Lynch C, Coble R, Mas E, Palsson O, Barrow DA, Mann JD. Targeted alteration of dietary n-3 and n-6 fatty acids for the treatment of chronic headaches: a randomized trial. Pain 2013;154:2441–2451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Ramsden CE, Mann JD, Faurot KR, Lynch C, Imam ST, MacIntosh BA, Hibbeln JR, Loewke J, Smith S, Coble R, Suchindran C, Gaylord SA. Low omega-6 vs. low omega-6 plus high omega-3 dietary intervention for chronic daily headache: protocol for a randomized clinical trial. Trials 2011;12:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Ramsden CE, Ringel A, Majchrzak-Hong SF, Yang J, Blanchard H, Zamora D, Loewke JD, Rapoport SI, Hibbeln JR, Davis JM, Hammock BD, Taha AY. Dietary linoleic acid-induced alterations in pro- and anti-nociceptive lipid autacoids: Implications for idiopathic pain syndromes? Mol. Pain 2016;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Ramsden CE, Yuan Z-X, Horowitz MS, Zamora D, Majchrzak-Hong SF, Muhlhausler BS, Taha AY, Makrides M, Gibson RA. Temperature and time-dependent effects of delayed blood processing on oxylipin concentrations in human plasma. Prostaglandins Leukot Essent Fatty Acids 2019;150:31–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Rose S, Marzi I. Mediators in polytrauma--pathophysiological significance and clinical relevance. Langenbecks Arch Surg 1998;383:199–208. [DOI] [PubMed] [Google Scholar]
- [44].Roux A, Muller L, Jackson SN, Post J, Baldwin K, Hoffer B, Balaban CD, Barbacci D, Schultz JA, Gouty S, Cox BM, Woods AS. Mass spectrometry imaging of rat brain lipid profile changes over time following traumatic brain injury. J. Neurosci. Methods 2016;272:19–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Rund KM, Nolte F, Doricic J, Greite R, Schott S, Lichtinghagen R, Gueler F, Schebb NH. Clinical blood sampling for oxylipin analysis - effect of storage and pneumatic tube transport of blood on free and total oxylipin profile in human plasma and serum. Analyst 2020;145:2378–2388. [DOI] [PubMed] [Google Scholar]
- [46].Schwab JM, Chiang N, Arita M, Serhan CN. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature 2007;447:869–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Seidl JNT, Pastorek NJ, Lillie R, Rosenblatt A, Troyanskaya M, Miller BI, Romesser J, Lippa S, Sim AH, Linck J. Factors related to satisfaction with life in veterans with mild traumatic brain injury. Rehabil Psychol 2015;60:335–343. [DOI] [PubMed] [Google Scholar]
- [48].Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature 2014;510:92–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Silverberg ND, Lange RT, Iverson GL. Concussion and Mild Traumatic Brain Injury: Definitions, Distinctions, and Diagnostic Criteria ∣ Neupsy Key. Concussion and Mild Traumatic Brain Injury: Definitions, Distinctions, and Diagnostic Criteria 2017. Available: https://neupsykey.com/concussion-and-mild-traumatic-brain-injury-definitions-distinctions-and-diagnostic-criteria/#T6-1. Accessed 4 Mar 2020.
- [50].Soares A de A, Louçana PMC, Nasi EP, Sousa KM de H, Sá OM de S, Silva-Néto RP. A double- blind, randomized, and placebo-controlled clinical trial with omega-3 polyunsaturated fatty acids (OPFA ɷ−3) for the prevention of migraine in chronic migraine patients using amitriptyline. Nutr Neurosci 2018;21:219–223. [DOI] [PubMed] [Google Scholar]
- [51].Solomon S. Chronic post-traumatic neck and head pain. Headache 2005;45:53–67. [DOI] [PubMed] [Google Scholar]
- [52].Sorokin AV, Domenichiello AF, Dey AK, Yuan Z-X, Goyal A, Rose SM, Playford MP, Ramsden CE, Mehta NN. Bioactive lipid mediator profiles in human psoriasis skin and blood. J. Invest. Dermatol 2018;138:1518–1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Theeler B, Lucas S, Riechers RG, Ruff RL. Post-traumatic headaches in civilians and military personnel: a comparative, clinical review. Headache 2013;53:881–900. [DOI] [PubMed] [Google Scholar]
- [54].Vane JR. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nature New Biol. 1971;231:232–235. [DOI] [PubMed] [Google Scholar]
- [55].Vargas BB, Dodick DW. Posttraumatic headache. Curr. Opin. Neurol 2012;25:284–289. [DOI] [PubMed] [Google Scholar]
- [56].Varma S, Janesko KL, Wisniewski SR, Bayir H, Adelson PD, Thomas NJ, Kochanek PM. F2-isoprostane and neuron-specific enolase in cerebrospinal fluid after severe traumatic brain injury in infants and children. J. Neurotrauma 2003;20:781–786. [DOI] [PubMed] [Google Scholar]
- [57].Wagner K, Vito S, Inceoglu B, Hammock BD. The role of long chain fatty acids and their epoxide metabolites in nociceptive signaling. Prostaglandins Other Lipid Mediat 2014;113–115:2–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Watanabe TK, Bell KR, Walker WC, Schomer K. Systematic review of interventions for post-traumatic headache. PM R 2012;4:129–140. [DOI] [PubMed] [Google Scholar]
- [59].Westcott JY, Murphy RC, Stenmark K. Eicosanoids in human ventricular cerebrospinal fluid following severe brain injury. Prostaglandins 1987;34:877–887. [DOI] [PubMed] [Google Scholar]
- [60].Xu Z-Z, Zhang L, Liu T, Park JY, Berta T, Yang R, Serhan CN, Ji R-R. Resolvins RvE1 and RvD1 attenuate inflammatory pain via central and peripheral actions. Nat. Med 2010;16:592–7, 1p following 597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Yi L, Shi S, Wang Y, Huang W, Xia Z, Xing Z, Peng W, Wang Z. Serum Metabolic Profiling Reveals Altered Metabolic Pathways in Patients with Post-traumatic Cognitive Impairments. Sci. Rep 2016;6:21320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Yu G-F, Jie Y-Q, Wu A, Huang Q, Dai W-M, Fan X-F. Increased plasma 8-iso-prostaglandin F2α concentration in severe human traumatic brain injury. Clin. Chim. Acta 2013;421:7–11. [DOI] [PubMed] [Google Scholar]
- [63].Yuan Z-X, Majchrzak-Hong S, Keyes GS, Iadarola MJ, Mannes AJ, Ramsden CE. Lipidomic profiling of targeted oxylipins with ultra-performance liquid chromatography-tandem mass spectrometry. Anal. Bioanal. Chem 2018;410:6009–6029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Zhang L, Terrando N, Xu Z-Z, Bang S, Jordt S-E, Maixner W, Serhan CN, Ji R-R. Distinct Analgesic Actions of DHA and DHA-Derived Specialized Pro-Resolving Mediators on Post-operative Pain After Bone Fracture in Mice. Front. Pharmacol 2018;9:412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Zimmer B, Angioni C, Osthues T, Toewe A, Thomas D, Pierre SC, Geisslinger G, Scholich K, Sisignano M. The oxidized linoleic acid metabolite 12,13-DiHOME mediates thermal hyperalgesia during inflammatory pain. Biochim. Biophys. Acta 2018;1863:669–678. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



