Oleoylethanolamide treatment reduces neurobehavioral deficits and brain pathology in a mouse model of Gulf War Illness

Utsav Joshi, James E Evans, Ross Joseph, Tanja Emmerich, Nicole Saltiel, Carlyn Lungmus, Sarah Oberlin, Heather Langlois, Joseph Ojo, Benoit Mouzon, Daniel Paris, Michael Mullan, Chao Jin, Nancy Klimas, Kimberly Sullivan, Fiona Crawford, Laila Abdullah, Utsav Joshi, James E Evans, Ross Joseph, Tanja Emmerich, Nicole Saltiel, Carlyn Lungmus, Sarah Oberlin, Heather Langlois, Joseph Ojo, Benoit Mouzon, Daniel Paris, Michael Mullan, Chao Jin, Nancy Klimas, Kimberly Sullivan, Fiona Crawford, Laila Abdullah

Abstract

There are nearly 250,000 Gulf War (GW) veterans who suffer from Gulf War Illness (GWI), a multi-symptom condition that remains untreatable. The main objective was to determine if targeting peroxisomal function could be of therapeutic value in GWI. We performed a pilot study that showed accumulation of very long chain fatty acids (VLCFA), which are metabolized in peroxisomes, in plasma from veterans with GWI. We then examined if targeting peroxisomal β-oxidation with oleoylethanolamide (OEA) restores these lipids to the normal levels and mitigates neuroinflammation and neurobehavioral deficits in a well-established mouse model of GWI. In GWI mice, treatment with OEA corresponded with cognitive benefits and reduced fatigue and disinhibition-like behavior in GWI mice. Biochemical and molecular analysis of the brain tissue showed reduced astroglia and microglia staining, decreased levels of chemokines and cytokines, and decreased NFκB phosphorylation. Treatment with OEA reduced accumulation of peroxisome specific VLCFA in the brains of GWI mice. These studies further support the translational value of targeting peroxisomes. We expect that OEA may be a potential therapy for treating neurobehavioral symptoms and the underlying lipid dysfunction and neuroinflammation associated with GWI. Oleoylethanolamide is available as a dietary supplement, making it appealing for human translational studies.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Plasma FFA profiles in veterans with GWI compared to GW control veterans. Mean ± SEM (n = 10 for controls and n = 12 for GWI). (A) When all VLCFA (C 22), were combined as one category, there was an overall increase in GWI compared to control GW veterans. (B) Quantification of lipid peroxidation product TBARS (n = 8 per group for these analyses only) in plasma showed that levels were significantly elevated in GWI compared to control veterans. (C) Profiling of FFA showed that several saturated (no double-bonds) and monounsaturated fatty acids (one double-bond) were decreased in the blood of veterans with GWI compared to controls. Pristanic acid was decreased in GWI compared to controls. The remaining long-chain fatty acids (14–21 carbons) and VLCFA species particularly omega-3 and omega-6 species, were significantly elevated in veterans with GWI compared to controls. *p ≤ 0.05.
Figure 2
Figure 2
The experimental paradigm. The experimental timeline details the timing of GW chemical administration, treatment with OEA, behavioral testing, and tissue collection.
Figure 3
Figure 3
OEA treatment improves cognitive function and reduces fatigue and disinhibition type behavior in GWI mice. Mean ± SEM of control (n = 12), GWI mice (n = 11), control + OEA (n = 12) and GWI + OEA (n = 12). (A) One month after OEA treatment (6-months post-exposure to GW chemicals), acquisition trials were conducted to train mice to escape into the TH. For GWI mice treated with OEA, total distance to the TH was smallest, particularly on days 1 and 2, indicative of learning. As expected, GWI mice on normal chow had the worst performance with the largest total distance to the TH. Control mice treated with OEA had a higher latency to find the TH compared to control mice on the normal diet on day 2 only. (B) For the probe trials, conducted at 2-months post-treatment (7-months post-exposure), OEA treated GWI mice had a similar frequency of visits to the TH as control mice on normal chow and on OEA but a higher frequency of visits than GWI mice. (C) The immobile time was significantly increased in GWI mice compared to controls and was reduced in OEA treated GWI mice. (D) There was no change in the average speed among the different groups. (E) An examination of the time in the open arms of the EPM showed an increase of disinhibition in GWI mice on normal chow compared to both control groups and OEA treated GWI mice. (F) Total time in the closed arms was decreased in GWI mice compared with the control groups and OEA treated GWI mice. *p ≤ 0.05.
Figure 4
Figure 4
Brain lipid profiles show that OEA significantly decreases the VLCFA in the brains of GWI mice. Mean ± SEM expressed as percent control of mice on normal chow (n = 4/5 per group). (A) Levels of VLCFA were increased in the brains of GWI mice compared to control mice. OEA treated GWI mice had similar levels as in control mice. However, control mice treated with OEA had significantly higher levels of VLCFA compared to control mice that received normal diet. (B) Quantification of lipid peroxidation product TBARS showed that levels were significantly elevated in GWI compared to control mice on normal chow but were lower in OEA treated GWI mice compared to all other groups. (C) Compared to controls, GWI mice had higher levels of several major omega-6 and omega-3 FA (FA20:4, FA22:4. FA22:5, FA24:5 and FA24:6). Treatment with OEA decreased the levels of these FA in GWI mice. *p ≤ 0.05.
Figure 5
Figure 5
OEA treatment reduced elevated astroglia activation in GWI mice. Confocal microscopy images showing staining of astroglia with GFAP (n = 4 per group, 4 serial sections for each animal). Scale bar 50 μm. (A) Images show 20x GFAP staining (red) and DAPI (blue) in the cortex and the DG of mice from all 4 treatment groups. (B) Images show 100x magnification of GFAP stained astroglia in the cortex of each study treatment group. (C) Quantification of GFAP staining from 20x images. There are significant increases in GFAP staining for both the cortex and the DG within the hippocampus of GWI compared to control mice. GWI mice treated with OEA had similar levels as control mice on normal chow. Control mice treated with OEA had higher levels than control mice on the normal diet. *p ≤ 0.05.
Figure 6
Figure 6
OEA treatment reduced microglia proliferation in GWI mice. Light microscopy images showing staining of microglia with IBA-1 (n = 4 per group, 4 serial section for each brain). Scale bar 50 μm. (A) Images show 20x Iba1 staining in the cortex and DG of mice from all 4 treatment groups. (B) Images show 100x magnification of Iba1 stained astroglia in the cortex of each study treatment group. (C) Quantification of Iba1 staining from 20x images. Levels of Iba1 staining within the DG was lower in OEA treated GWI mice compared to GWI mice on normal chow. Levels of Iba1 in GWI mice were higher in the DG compared to control mice. There were no differences in the cortices of GWI mice compared to control or OEA treated GWI mice *p ≤ 0.05.
Figure 7
Figure 7
Levels of phosphorylated NFκB and STAT3 were reduced by OEA treatment in GWI mice. Mean ± SEM (shown as arbitrary units, n = 4 per group). (A,B) The ratio of p-P65/P65 was elevated in GWI mice at 11-months post-exposure. At this time-point, which corresponded with 6-months of OEA treatment, GWI mice having OEA treatment had a significantly lower ratio compared to GWI mice on normal chow. Control mice treated with OEA had non-significantly lower ratios compared to control mice on the regular diet. (C,D) Similarly, p-STAT3/Actin levels were significantly elevated in GWI compared to control mice and were also lower in OEA treated GWI mice compared to those on normal chow. Levels of STAT3 were also elevated in control mice treated with OEA compared to control mice on the normal diet. (E) Among the cytokines examined in the brain, IFN-γ, IL-6, and IL-1β were lower in OEA treated GWI mice compared to GWI mice on normal chow. Mice with GWI had elevated levels of these pro-inflammatory cytokines compared to control mice. These cytokines did not differ between control mice treated with OEA compared to control mice on the normal diet. (F) Treatment with OEA decreased the levels of CCR2 and CCL2 in the brain of GWI mice compared to GWI mice on normal chow. *p ≤ 0.05.
Figure 8
Figure 8
Gulf War chemicals dysregulate peroxisomal lipid metabolism in astroglia, contributing to microglia activation and inflammation which corresponds with neurobehavioral deficits. Our working hypothesis is that GW chemicals affect peroxisomal lipid metabolism in the astrocytes since astroglia activation occurs earlier than microglia activation in this GWI mouse model. We also propose that the failure of astroglia to perform its neuroprotective function contributes to the subsequent activation of microglia and neuroinflammation. Targeting peroxisomes with OEA is expected to reduce astroglia activation which corresponds with normalization of peroxisomal lipid metabolism. We also propose that OEA may also reduce NFκB activation in microglia, thereby reducing neuroinflammation. In another scenario, OEA may undergo degradation by FAAH and oxygenation by other enzymes, generating additional bioactive metabolites which may target the cannabinoid receptors and also promote anti-inflammatory responses via synergistic activation of NFκB.

References

    1. White RF, et al. Recent research on Gulf War illness and other health problems in veterans of the 1991 Gulf War: Effects of toxicant exposures during deployment. Cortex. 2016;74:449–475. doi: 10.1016/j.cortex.2015.08.022.
    1. Rayhan Rakib U., Stevens Benson W., Timbol Christian R., Adewuyi Oluwatoyin, Walitt Brian, VanMeter John W., Baraniuk James N. Increased Brain White Matter Axial Diffusivity Associated with Fatigue, Pain and Hyperalgesia in Gulf War Illness. PLoS ONE. 2013;8(3):e58493. doi: 10.1371/journal.pone.0058493.
    1. Binns, J. et al. Gulf War Illness and the Health of Gulf War Veterans: Scientific Findings and Recommendations. 1–465 (2008).
    1. Terry AV, et al. Chronic, intermittent exposure to chlorpyrifos in rats: protracted effects on axonal transport, neurotrophin receptors, cholinergic markers, and information processing. J. Pharmacol. Exp. Ther. 2007;322:1117–1128. doi: 10.1124/jpet.107.125625.
    1. Abdullah L, et al. Lipidomic profiling of phosphocholine containing brain lipids in mice with sensorimotor deficits and anxiety-like features after exposure to gulf war agents. NeuroMolecular Med. 2012;14:349–361. doi: 10.1007/s12017-012-8192-z.
    1. Abdullah L, et al. Proteomic CNS profile of delayed cognitive impairment in mice exposed to Gulf War agents. Neuro Molecular Med. 2011;13:275–288. doi: 10.1007/s12017-011-8160-z.
    1. Zakirova Z, et al. A chronic longitudinal characterization of neurobehavioral and neuropathological cognitive impairment in a mouse model of Gulf War agent exposure. Front. Integr. Neurosci. 2015;9:71.
    1. Abdullah L, et al. Translational potential of long-term decreases in mitochondrial lipids in a mouse model of Gulf War Illness. Toxicology. 2016;372:22–33. doi: 10.1016/j.tox.2016.10.012.
    1. Parihar VK, Hattiangady B, Shuai B, Shetty AK. Mood and memory deficits in a model of Gulf War illness are linked with reduced neurogenesis, partial neuron loss, and mild inflammation in the hippocampus. Neuropsychopharmacology. 2013;38:2348–62. doi: 10.1038/npp.2013.158.
    1. Abdullah L, et al. Chronic elevation of phosphocholine containing lipids in mice exposed to Gulf War agents pyridostigmine bromide and permethrin. Neurotoxicol. Teratol. 2013;40:74–84. doi: 10.1016/j.ntt.2013.10.002.
    1. Emmerich Tanja, Zakirova Zuchra, Klimas Nancy, Sullivan Kimberly, Shetty Ashok K., Evans James E., Ait-Ghezala Ghania, Laco Gary S., Hattiangady Bharathi, Shetty Geetha A., Mullan Michael, Crynen Gogce, Abdullah Laila, Crawford Fiona. Phospholipid profiling of plasma from GW veterans and rodent models to identify potential biomarkers of Gulf War Illness. PLOS ONE. 2017;12(4):e0176634. doi: 10.1371/journal.pone.0176634.
    1. Ferdinandusse S, et al. Identification of the peroxisomal beta-oxidation enzymes involved in the biosynthesis of docosahexaenoic acid. J. Lipid Res. 2001;42:1987–95.
    1. Casteels M, Foulon V, Mannaerts GP, Van Veldhoven PP. Alpha-oxidation of 3-methyl-substituted fatty acids and its thiamine dependence. European Journal of Biochemistry. 2003;270:1619–1627. doi: 10.1046/j.1432-1033.2003.03534.x.
    1. Watkins PA. Very-long-chain acyl-CoA synthetases. Journal of Biological Chemistry. 2008;283:1773–1777. doi: 10.1074/jbc.R700037200.
    1. Mashek DG, Greenberg AS. Serum TAG analysis differentiates between genetic and obesity-associated NAFLD. Diabetes. 2014;63:42–44. doi: 10.2337/db13-1500.
    1. Terlecky SR, Terlecky LJ, Giordano CR. Peroxisomes, oxidative stress, and inflammation. World J. Biol. Chem. 2012;3:93–7. doi: 10.4331/wjbc.v3.i5.93.
    1. Ivashchenko O, et al. Intraperoxisomal redox balance in mammalian cells: oxidative stress and interorganellar cross-talk. Mol. Biol. Cell. 2011;22:1440–1451. doi: 10.1091/mbc.e10-11-0919.
    1. Koepke JI, Wood CS, Terlecky LJ, Walton PA, Terlecky SR. Progeric effects of catalase inactivation in human cells. Toxicol. Appl. Pharmacol. 2008;232:99–108. doi: 10.1016/j.taap.2008.06.004.
    1. Shetty GA, et al. Chronic Oxidative Stress, Mitochondrial Dysfunction, Nrf2 Activation and Inflammation in the Hippocampus Accompany Heightened Systemic Inflammation and Oxidative Stress in an Animal Model of Gulf War Illness. Front. Mol. Neurosci. 2017;10:1–20. doi: 10.3389/fnmol.2017.00182.
    1. Khaiboullina SF, et al. Cytokine expression provides clues to the pathophysiology of Gulf War illness and myalgic encephalomyelitis. Cytokine. 2015;72:1–8. doi: 10.1016/j.cyto.2014.11.019.
    1. Broderick G, et al. Exploring the diagnostic potential of immune biomarker coexpression in gulf war illness. Methods Mol. Biol. 2012;934:145–164. doi: 10.1007/978-1-62703-071-7_8.
    1. Vojdani A, Thrasher JD. Cellular and humoral immune abnormalities in Gulf War veterans. Environ. Health Perspect. 2004;112:840–846. doi: 10.1289/ehp.6881.
    1. Zhang Q, et al. Changes in immune parameters seen in Gulf War veterans but not in civilians with chronic fatigue syndrome. Clin. Diagn. Lab. Immunol. 1999;6:6–13.
    1. Bordet R, et al. PPAR: a new pharmacological target for neuroprotection in stroke and neurodegenerative diseases. Biochem Soc Trans. 2006;34:1341–1346. doi: 10.1042/BST0341341.
    1. Jung TW, et al. Rosiglitazone protects human neuroblastoma SH-SY5Y cells against MPP+ induced cytotoxicity via inhibition of mitochondrial dysfunction and ROS production. J. Neurol. Sci. 2007;253:53–60. doi: 10.1016/j.jns.2006.11.020.
    1. Gray E, Ginty M, Kemp K, Scolding N, Wilkins A. Peroxisome proliferator-activated receptor-α agonists protect cortical neurons from inflammatory mediators and improve peroxisomal function. Eur. J. Neurosci. 2011;33:1421–1432. doi: 10.1111/j.1460-9568.2011.07637.x.
    1. Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors and. Proc. Natl. Acad. Sci. 1997;94:4312–4317. doi: 10.1073/pnas.94.9.4312.
    1. Lehmann JM, Lenhard JM, Oliver BB, Ringold GM, Kliewer SA. Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti-inflammatory drugs. J. Biol. Chem. 1997;272:3406–10. doi: 10.1074/jbc.272.6.3406.
    1. Cunard R, et al. WY14,643, a PPAR alpha ligand, has profound effects on immune responses in vivo. J. Immunol. 2002;169:6806–6812. doi: 10.4049/jimmunol.169.12.6806.
    1. Jones DC, Ding X, Daynes R. a. Nuclear receptor peroxisome proliferator-activated receptor alpha (PPARalpha) is expressed in resting murine lymphocytes. The PPARalpha in T and B lymphocytes is both transactivation and transrepression competent. J. Biol. Chem. 2002;277:6838–45. doi: 10.1074/jbc.M106908200.
    1. Dasgupta S, Roy A, Jana M, Hartley DM, Pahan K. Gemfibrozil ameliorates relapsing-remitting experimental autoimmune encephalomyelitis independent of peroxisome proliferator-activated receptor-alpha. Mol. Pharmacol. 2007;72:934–946. doi: 10.1124/mol.106.033787.
    1. Gocke AR, et al. Transcriptional modulation of the immune response by peroxisome proliferator-activated receptor-{alpha} agonists in autoimmune disease. J. Immunol. 2009;182:4479–87. doi: 10.4049/jimmunol.0713927.
    1. Zakirova Zuchra, Tweed Miles, Crynen Gogce, Reed Jon, Abdullah Laila, Nissanka Nadee, Mullan Myles, Mullan Michael J., Mathura Venkatarajan, Crawford Fiona, Ait-Ghezala Ghania. Gulf War Agent Exposure Causes Impairment of Long-Term Memory Formation and Neuropathological Changes in a Mouse Model of Gulf War Illness. PLOS ONE. 2015;10(3):e0119579. doi: 10.1371/journal.pone.0119579.
    1. Duan, F. F., Guo, Y., Li, J. W. & Yuan, K. Antifatigue Effect of Luteolin-6-C-Neohesperidoside on Oxidative Stress Injury Induced by Forced Swimming of Rats through Modulation of Nrf2/ARE SignalingPathways. Oxid. Med. Cell. Longev. 2017 (2017).
    1. Xu M, Liang R, Li Y, Wang J. Anti-fatigue effects of dietary nucleotides in mice. Food Nutr. Res. 2017;61:1334485. doi: 10.1080/16546628.2017.1334485.
    1. Gadea A, López-Colomé AM. Glial transporters for glutamate, glycine, and GABA: II. GABA transporters. Journal of Neuroscience Research. 2001;63:461–468. doi: 10.1002/jnr.1040.
    1. Kacem K, Lacombe P, Seylaz J, Bonvento G. Structural organization of the perivascular astrocyte endfeet and their relationship with the endothelial glucose transporter: A confocal microscopy study. Glia. 1998;23:1–10. doi: 10.1002/(SICI)1098-1136(199805)23:1<1::AID-GLIA1>;2-B.
    1. Gehrmann J, Matsumoto Y, Kreutzberg GW. Microglia: Intrinsic immuneffector cell of the brain. Brain Research Reviews. 1995;20:269–287. doi: 10.1016/0165-0173(94)00015-H.
    1. Kreutzberg GW. Microglia: A sensor for pathological events in the CNS. Trends in Neurosciences. 1996;19:312–318. doi: 10.1016/0166-2236(96)10049-7.
    1. Jimenez F, et al. CCR2 plays a critical role in dendritic cell maturation: possible role of CCL2 and NF-kappa B. J. Immunol. 2010;184:5571–81. doi: 10.4049/jimmunol.0803494.
    1. Chen K, et al. Signal relay by CC chemokine receptor 2 (CCR2) and formylpeptide receptor 2 (Fpr2) in the recruitment of monocyte-derived dendritic cells in allergic airway inflammation. J. Biol. Chem. 2013;288:16262–16273. doi: 10.1074/jbc.M113.450635.
    1. Macanas-Pirard Patricia, Quezada Thomas, Navarrete Leonardo, Broekhuizen Richard, Leisewitz Andrea, Nervi Bruno, Ramírez Pablo A. The CCL2/CCR2 Axis Affects Transmigration and Proliferation but Not Resistance to Chemotherapy of Acute Myeloid Leukemia Cells. PLOS ONE. 2017;12(1):e0168888. doi: 10.1371/journal.pone.0168888.
    1. Maule AL, et al. Meta-analysis of self-reported health symptoms in 1990–1991 Gulf War and Gulf War-era veterans. BMJ Open. 2018;8:e016086. doi: 10.1136/bmjopen-2017-016086.
    1. Sullivan K, et al. Cognitive functioning in treatment-seeking Gulf War veterans: Pyridostigmine bromide use and PTSD. J. Psychopathol. Behav. Assess. 2003;25:95–103. doi: 10.1023/A:1023342915425.
    1. Anger WK, et al. Neurobehavioral deficits in Persian Gulf veterans: Evidence from a population-based study. J. Int. Neuropsychol. Soc. 1999;5:203–212. doi: 10.1017/S1355617799533031.
    1. Sillanpaa MC, et al. Gulf War veterans: A neuropsychological examination. J. Clin. Exp. Neuropsychol. 1997;19:211–219. doi: 10.1080/01688639708403852.
    1. Binder LM, et al. Subjective cognitive complaints, affective distress, and objective cognitive performance in Persian Gulf War veterans. Arch. Clin. Neuropsychol. 1999;14:531–6. doi: 10.1093/arclin/14.6.531.
    1. Bunegin L, Mitzel HC, Miller CS, Gelineau JF, Tolstykh GP. Cognitive performance and cerebrohemodynamics associated with the Persian Gulf Syndrome. Toxicol. Ind. Health. 2001;17:128–137. doi: 10.1191/0748233701th100oa.
    1. Lange G, Van Niekerk A, Meyer BJ. Detection of an artifact on lumbar SPECT. Clin. Nucl. Med. 2001;26:446–448. doi: 10.1097/00003072-200105000-00014.
    1. Storzbach D, et al. Psychological differences between veterans with and without Gulf War unexplained symptoms. Psychosom. Med. 2000;62:726–735. doi: 10.1097/00006842-200009000-00017.
    1. Menon S, et al. A longitudinal study of anticardiolipin antibody levels and cognitive functioning in systemic lupus erythematosus. Arthritis Rheum. 1999;42:735–741. doi: 10.1002/1529-0131(199904)42:4<735::AID-ANR17>;2-L.
    1. Vythilingam M, et al. Smaller head of the hippocampus in Gulf War-related posttraumatic stress disorder. Psychiatry Res. - Neuroimaging. 2005;139:89–99. doi: 10.1016/j.pscychresns.2005.04.003.
    1. Weiner, M. W. Magnetic Resonance and Spectroscopy of the Human Brain in Gulf War Illness. 1–11 (2005).
    1. Koo BB, et al. Corticosterone potentiates DFP-induced neuroinflammation and affects high-order diffusion imaging in a rat model of Gulf War Illness. Brain. Behav. Immun. 2018;67:42–46. doi: 10.1016/j.bbi.2017.08.003.
    1. Terry AV. Functional Consequences of Repeated Organophosphate Exposure: Potential Non-Cholinergic Mechanisms. Pharmacol Ther. 2012;v:265–275.
    1. Campolongo P, et al. Fat-induced satiety factor oleoylethanolamide enhances memory consolidation. Proc. Natl. Acad. Sci. USA. 2009;106:8027–8031. doi: 10.1073/pnas.0903038106.
    1. Yang LC, et al. Chronic oleoylethanolamide treatment improves spatial cognitive deficits through enhancing hippocampal neurogenesis after transient focal cerebral ischemia. Biochem. Pharmacol. 2015;94:257–269. doi: 10.1016/j.bcp.2015.02.012.
    1. Plaza-Zabala A, et al. Effects of the endogenous PPAR-alpha agonist, oleoylethanolamide on MDMA-induced cognitive deficits in mice. Synapse. 2010;64:379–389. doi: 10.1002/syn.20733.
    1. Bannerman DM, et al. Regional dissociations within the hippocampus - Memory and anxiety. Neuroscience and Biobehavioral Reviews. 2004;28:273–283. doi: 10.1016/j.neubiorev.2004.03.004.
    1. Da Cruz e Alves-de-Moraes LB, Ribeiro-Paes JT, Longo BM, Ferrazoli EG, De Andrade TGCS. Effect of the bone marrow cell transplantation on elevated plus-maze performance in hippocampal-injured mice. Behav. Brain Res. 2013;248:32–40. doi: 10.1016/j.bbr.2013.03.042.
    1. Rayhan Rakib U., Stevens Benson W., Raksit Megna P., Ripple Joshua A., Timbol Christian R., Adewuyi Oluwatoyin, VanMeter John W., Baraniuk James N. Exercise Challenge in Gulf War Illness Reveals Two Subgroups with Altered Brain Structure and Function. PLoS ONE. 2013;8(6):e63903. doi: 10.1371/journal.pone.0063903.
    1. Abou-Donia MB, et al. Screening for novel central nervous system biomarkers in veterans with Gulf War Illness. Neurotoxicol. Teratol. 2017;61:36–46. doi: 10.1016/j.ntt.2017.03.002.
    1. McKimmie CS, Graham GJ. Astrocytes modulate the chemokine network in a pathogen-specific manner. Biochem. Biophys. Res. Commun. 2010;394:1006–1011. doi: 10.1016/j.bbrc.2010.03.111.
    1. Ramesh, G., Maclean, A. G. & Philipp, M. T. Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration, and neuropathic pain. Mediators Inflamm. 2013 (2013).
    1. Conductier G, Blondeau N, Guyon A, Nahon JL, Rovère C. The role of monocyte chemoattractant protein MCP1/CCL2 in neuroinflammatory diseases. J. Neuroimmunol. 2010;224:93–100. doi: 10.1016/j.jneuroim.2010.05.010.
    1. Selenica, M. L. B. et al. Diverse activation of microglia by chemokine (C-C motif) ligand 2 overexpression in brain. J. Neuroinflammation10 (2013).
    1. Parillaud, V. R. et al. Analysis of monocyte infiltration in MPTP mice reveals that microglial CX3CR1 protects against neurotoxic over-induction of monocyte-attracting CCL2 by astrocytes. J. Neuroinflammation14 (2017).
    1. Parkitny L, Middleton S, Baker K, Younger J. Evidence for abnormal cytokine expression in Gulf War Illness: A preliminary analysis of daily immune monitoring data. BMC Immunol. 2015;16:57. doi: 10.1186/s12865-015-0122-z.
    1. Craddock Travis J. A., Del Rosario Ryan R., Rice Mark, Zysman Joel P., Fletcher Mary Ann, Klimas Nancy G., Broderick Gordon. Achieving Remission in Gulf War Illness: A Simulation-Based Approach to Treatment Design. PLOS ONE. 2015;10(7):e0132774. doi: 10.1371/journal.pone.0132774.
    1. Sagar, D. R. et al. Dynamic regulation of the endocannabinoid system: Implications for analgesia. Molecular Pain5 (2009).
    1. Watt MJ, Southgate RJ, Holmes AG, Febbraio MA. Suppression of plasma free fatty acids upregulates peroxisome proliferator-activated receptor (PPAR) alpha and delta and PPAR coactivator 1alpha in human skeletal muscle, but not lipid regulatory genes. J. Mol. Endocrinol. 2004;33:533–44. doi: 10.1677/jme.1.01499.
    1. Edvardsson U, et al. PPARalpha activation increases triglyceride mass and adipose differentiation-related protein in hepatocytes. J. Lipid Res. 2006;47:329–40. doi: 10.1194/jlr.M500203-JLR200.
    1. Garcia-Roves P, et al. Raising plasma fatty acid concentration induces increased biogenesis of mitochondria in skeletal muscle. Proc. Natl. Acad. Sci. 2007;104:10709–10713. doi: 10.1073/pnas.0704024104.
    1. Basaure P, et al. Postnatal chlorpyrifos exposure and apolipoprotein E (APOE) genotype di ff erentially a ff ect cholinergic expression and developmental parameters in transgenic mice. Food Chem. Toxicol. 2018;118:42–52. doi: 10.1016/j.fct.2018.04.065.
    1. Peris-Sampedro F, et al. Apolipoprotein E (APOE) genotype and the pesticide chlorpyrifos modulate attention, motivation and impulsivity in female mice in the 5-choice serial reaction time task. Food Chem. Toxicol. 2016;92:224–235. doi: 10.1016/j.fct.2016.03.029.
    1. Torres-Platas, S. G. et al. Morphometric characterization of microglial phenotypes in human cerebral cortex. J. Neuroinflammation11 (2014).
    1. Arlicot N, et al. Evaluation of CLINDE as potent translocator protein (18 kDa) SPECT radiotracer reflecting the degree of neuroinflammation in a rat model of microglial activation. Eur. J. Nucl. Med. Mol. Imaging. 2008;35:2203–2211. doi: 10.1007/s00259-008-0834-x.
    1. Winkeler A, et al. The translocator protein ligand [18F]DPA-714 images glioma and activated microglia in vivo. Eur. J. Nucl. Med. Mol. Imaging. 2012;39:811–823. doi: 10.1007/s00259-011-2041-4.
    1. Arlicot N, et al. Detection and quantification of remote microglial activation in rodent models of focal ischaemia using the TSPO radioligand CLINDE. Eur. J. Nucl. Med. Mol. Imaging. 2010;37:2371–2380. doi: 10.1007/s00259-010-1598-7.
    1. Folch J, Lees M, Sloane Stanley GH. A simple method for the isolation and purification of total lipides from animal tissues. J. Biol. Chem. 1957;226:497–509.
    1. Flygt Johanna, Ruscher Karsten, Norberg Amanda, Mir Anis, Gram Hermann, Clausen Fredrik, Marklund Niklas. Neutralization of Interleukin-1β following Diffuse Traumatic Brain Injury in the Mouse Attenuates the Loss of Mature Oligodendrocytes. Journal of Neurotrauma. 2018;35(23):2837–2849. doi: 10.1089/neu.2018.5660.
    1. Boldrini M, et al. Human Hippocampal Neurogenesis Persists throughout Aging. Cell Stem Cell. 2018;22:589–599.e5. doi: 10.1016/j.stem.2018.03.015.
    1. Sorrells SF, et al. Human hippocampal neurogenesis drops sharply in children to undetectable levels in adults. Nature. 2018;555:377–381. doi: 10.1038/nature25975.
    1. Can, A. et al. The mouse forced swim test. J. Vis. Exp. e3638 10.3791/3638 (2012).
    1. Zhang Z, et al. NLRP3 inflammasome activation mediates fatigue-like behaviors in mice via neuroinflammation. Neuroscience. 2017;358:115–123. doi: 10.1016/j.neuroscience.2017.06.048.

Source: PubMed

3
Tilaa