Elsevier

Brain, Behavior, and Immunity

Volume 76, February 2019, Pages 17-27
Brain, Behavior, and Immunity

Dietary n-3 long chain PUFA supplementation promotes a pro-resolving oxylipin profile in the brain

https://doi.org/10.1016/j.bbi.2018.07.025Get rights and content

Highlights

  • Dietary n-3 LC-PUFA supplementation modulates hippocampal oxylipins profile.

  • Dietary n-3 LC-PUFA supplementation decreases hippocampal pro-inflammatory cytokines induced by LPS.

  • Oxylipin profile depending on dietary intake could orchestrate inflammatory response to LPS.

Abstract

The brain is highly enriched in long chain polyunsaturated fatty acids (LC-PUFAs) that display immunomodulatory properties in the brain. At the periphery, the modulation of inflammation by LC-PUFAs occurs through lipid mediators called oxylipins which have anti-inflammatory and pro-resolving activities when derived from n-3 LC-PUFAs and pro-inflammatory activities when derived from n-6 LC-PUFAs. However, whether a diet rich in LC-PUFAs modulates oxylipins and neuroinflammation in the brain has been poorly investigated. In this study, the effect of a dietary n-3 LC-PUFA supplementation on oxylipin profile and neuroinflammation in the brain was analyzed. Mice were given diets deficient or supplemented in n-3 LC-PUFAs for a 2-month period starting at post-natal day 21, followed by a peripheral administration of lipopolysaccharide (LPS) at adulthood. We first showed that dietary n-3 LC-PUFA supplementation induced n-3 LC-PUFA enrichment in the hippocampus and subsequently an increase in n-3 PUFA-derived oxylipins and a decrease in n-6 PUFA-derived oxylipins. In response to LPS, n-3 LC-PUFA deficient mice presented a pro-inflammatory oxylipin profile whereas n-3 LC-PUFA supplemented mice displayed an anti-inflammatory oxylipin profile in the hippocampus. Accordingly, the expression of cyclooxygenase-2 and 5-lipoxygenase, the enzymes implicated in pro- and anti-inflammatory oxylipin synthesis, was induced by LPS in both diets. In addition, LPS-induced pro-inflammatory cytokine increase was reduced by dietary n-3 LC-PUFA supplementation. These results indicate that brain n-3 LC-PUFAs increase by dietary means and promote the synthesis of anti-inflammatory derived bioactive oxylipins. As neuroinflammation plays a key role in all brain injuries and many neurodegenerative disorders, the present data suggest that dietary habits may be an important regulator of brain cytokine production in these contexts.

Introduction

The brain is highly enriched in long chain polyunsaturated fatty acids (LC-PUFAs) that are essential bioactive compounds with a large range of physiological roles such as cerebral plasticity, cell survival and neuroinflammation (Bazinet and Laye, 2014, Laye et al., 2018). Docosahexaenoic acid (DHA, 22:6 n-3) and arachidonic acid (AA, 20:4 n-6) are respectively the major n-3 and n-6 PUFAs in the brain. Despite the fact that DHA is poorly synthesized from its dietary precursor α-linolenic acid (ALA) (<1%) (Plourde and Cunnane, 2007), ALA-derived DHA has been recently reported to be sufficient for brain DHA supply (Domenichiello et al., 2014). However, preformed DHA can be provided through the diet to increase DHA levels in the brain (Orr et al., 2013, Joffre et al., 2016). N-3 LC-PUFA supplementation is the most efficient way to increase DHA levels and decrease AA levels in the brain (Murthy et al., 2002, Hiratsuka et al., 2009, Orr et al., 2010, Moranis et al., 2012, Fan et al., 2016, Lacombe et al., 2017).

N-3 LC-PUFAs exert protective actions against inflammation in the brain (Lonergan et al., 2004, Li et al., 2015, Shi et al., 2016, Dong et al., 2017, Fourrier et al., 2017). In vivo, fish oil supplementation providing n-3 LC-PUFAs, including DHA and eicosapentaenoic acid (EPA), is associated with a decreased cerebral expression of the pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β, induced by pro-inflammatory stimuli such as lipopolysaccharide (LPS, the gram-negative bacteria endotoxin), IL-1β (Dong et al., 2017) or aging (Labrousse et al., 2012, Orr et al., 2013, Dehkordi et al., 2015, Shi et al., 2016). In vitro data suggests that the anti-inflammatory activity of DHA in the brain is mediated through microglia, the brain’s innate immune cells (De Smedt-Peyrusse et al., 2008, Antonietta Ajmone-Cat et al., 2012, Pettit et al., 2013, Chang et al., 2015, Fourrier et al., 2017). Recent studies indicate that the mechanisms underlying the anti-inflammatory effect of DHA and EPA in the brain involve n-3 LC-PUFA derived bioactive mediators, called oxylipins, which promote the resolution of inflammation (Serhan et al., 2000, Levy et al., 2001, Hong et al., 2003, Serhan et al., 2015, Rey et al., 2016).

LC-PUFAs are converted by cyclooxygenase (COX), lipoxygenases (LOX) and cytochrome P450 (CYP) into oxylipins (Fig. 1). Several studies have reported their pro or anti-inflammatory activities in the brain (Orr et al., 2013, Rey et al., 2016, Laye et al., 2018). Oxylipins derived from DHA and EPA include anti-inflammatory and pro-resolving metabolites such as eicosanoids, resolvins, protectin and maresin (Bazan, 2009, Serhan et al., 2011). Oxylipins derived from AA and its precursor linoleic acid (LA) are mostly pro-inflammatory compounds and include prostaglandins (PG), leukotrienes (LT) or thromboxanes (Tx) and hydroxyoctadecadienoic acids (HODE) (Fig. 1) (Calder, 2006). Oxylipin levels are tightly regulated by inflammatory stimuli or insults in peripheral tissues and immune cells (Yang et al., 2009, Balvers et al., 2012a, Willenberg et al., 2016). In the brain, n-3 and n-6 derived oxylipins are produced during injury or ischemic conditions, including from microglia (Farias et al., 2008). The expression of oxylipin’s synthesizing enzymes is also tightly regulated by inflammatory stimuli (Rosenberger et al., 2004, Birnie et al., 2013, Taha et al., 2017), including in the brain (Laye et al., 2018). Administration of pro-inflammatory cytokines or LPS induces COX-2 expression in neuronal and endothelial cells (Cao et al., 1996, Nadjar et al., 2005, Rummel et al., 2006). COX-2, 5-LOX, 15-LOX and several isoforms of CYP expression are also induced in the hippocampus and cortex of rodents after traumatic brain injury (Birnie et al., 2013). A PUFA dietary intervention modulates both cellular levels of PUFAs and oxylipins. As a result, n-3 LC-PUFA supplementation increases oxylipins derived from EPA and DHA and decreases compounds derived from AA (Balvers et al., 2012b, Hashimoto et al., 2015). Conversely, dietary n-6 PUFA supplementation increases AA-derived oxylipins and decreases oxylipins derived from EPA (Taha et al., 2016). Changes of oxylipins levels triggered by dietary n-6 and n-3 PUFA supply could be linked to the regulation of the enzymes COX-2 and 15-LOX (Rao et al., 2007, Kim et al., 2011, Taha et al., 2017). However, the influence of dietary n-3 LC-PUFAs on the conversion enzymes and oxylipin profiles in specific brain regions has been poorly addressed. N-3 LC-PUFAs are promising molecules due to their modulation of brain inflammation (Lonergan et al., 2004, Li et al., 2015, Shi et al., 2016, Dong et al., 2017, Fourrier et al., 2017). Thus, we explored whether n-3 LC-PUFA dietary supplementation provides protection in an LPS model of inflammation through differential production of oxylipins.

We hypothesized that inflammatory stimuli can differently regulate n-3 LC-PUFA derived-oxylipins production in the brain, dependent on the dietary n-3 LC-PUFA supply. To address this, we compared the oxylipin profile in the hippocampus of LPS treated mice fed with isocaloric diets with identical ALA content but supplemented or deficient in n-3 LC-PUFAs.

Section snippets

Animals and treatment

All experiments were performed on male C57Bl6/J mice obtained from Janvier Labs at post-natal day 21 (Le Genest-Saint-Isle, France). Mice were maintained under standard housing conditions on corncob litter in a temperature (23 ± 1 °C) and humidity (40%) controlled animal room with a 12 h light/dark cycle (07–19 h), with ad libitum access to food and water. A total of 88 mice were used in this study. Half of the mice were fed with a diet enriched with n-3 LC-PUFA and the other half with a diet

Dietary n-3 LC-PUFA supplementation modifies hippocampal fatty acid composition

We first evaluated the impact of either diet on the hippocampal fatty acid composition (Table 4). Total n-3 PUFA and n-6 PUFA levels were significantly different in the hippocampus of the n-3 LC-PUFA supplemented group as compared to the n-3 LC-PUFA deficient group (DHA: F(1,12) = 25.22, p < 0.001, EPA: F(1,12) = 305.3, p < 0.0001, n-3 DPA: F(1,12) = 18.27, p < 0.001, AA: (F(1,12) = 211.1, p < 0.0001, n-6 docosapentaenoic acid (DPA): F(1,12) = 192.6, p < 0.0001, adrenic acid: F(1,12) = 174.4,

Discussion

In this work, we provide evidence that dietary n-3 LC-PUFA supplementation modulates hippocampal oxylipins and alters LPS-induced pro-inflammatory cytokine expression in the hippocampus, suggesting that n-3 LC-PUFAs could promote the resolution of hippocampal neuroinflammation through the release of oxylipins.

In the n-3 LC-PUFA supplemented diet, the amount of EPA and DHA corresponds to a daily intake of 40 mg/day/mouse (26.5 mg EPA and 14 mg DHA per mouse). To extrapolate to humans (Nair and

Acknowledgement

We thank C.Tridon, S. Delbary and B. Péré for taking care of the mice and Genotoul Plateforme for oxylipin analyses.

Funding source

This work was supported by INRA, ANRT, ACTIA (Association Nationale de la Recherche et de la Technologie), Idex Bordeaux University, ANR MoodFood, Fondation pour la Recherche Médicale (FRM) and Société Française de Nutrition (SFN). J.C. Delpech is the recipient of a post-doctoral fellow from Région Aquitaine. A.D. Greenhalgh is the recipient of the Agreenskills post-doctoral grant support. The funders had no role in study design, data collection and analysis, decision to publish, or preparation

Declaration of interest

None

References (94)

  • S.E. Farias et al.

    Formation of eicosanoids, E2/D2 isoprostanes, and docosanoids following decapitation-induced ischemia, measured in high-energy-microwaved rat brain

    J. Lipid Res.

    (2008)
  • M. Gabbs et al.

    Advances in our understanding of oxylipins derived from dietary PUFAs

    Adv. Nutr.

    (2015)
  • M. Hashimoto et al.

    n-3 fatty acids effectively improve the reference memory-related learning ability associated with increased brain docosahexaenoic acid-derived docosanoids in aged rats

    BBA

    (2015)
  • S. Hong et al.

    Novel docosatrienes and 17S-resolvins generated from docosahexaenoic acid in murine brain, human blood, and glial cells. Autacoids in anti-inflammation

    J. Biol. Chem.

    (2003)
  • J.J. Iliff et al.

    Epoxyeicosanoid signaling in CNS function and disease

    Prostaglandins Other Lipid Mediat.

    (2010)
  • C. Joffre et al.

    Modulation of brain PUFA content in different experimental models of mice

    Prostaglandins Leukot. Essent. Fatty Acids

    (2016)
  • H.W. Kim et al.

    Dietary n-6 PUFA deprivation downregulates arachidonate but upregulates docosahexaenoate metabolizing enzymes in rat brain

    BBA

    (2011)
  • R.J.S. Lacombe et al.

    Compound-specific isotope analysis resolves the dietary origin of docosahexaenoic acid in the mouse brain

    J. Lipid Res.

    (2017)
  • S. Laye

    Polyunsaturated fatty acids, neuroinflammation and well being

    Prostaglandins Leukot. Essent. Fatty Acids

    (2010)
  • S. Laye et al.

    Peripheral administration of lipopolysaccharide induces the expression of cytokine transcripts in the brain and pituitary of mice

    Brain Res. Mol. Brain Res.

    (1994)
  • P. Le Faouder et al.

    LC-MS/MS method for rapid and concomitant quantification of pro-inflammatory and pro-resolving polyunsaturated fatty acid metabolites

    J. Chromatogr. B Analyt. Technol. Biomed. Life Sci.

    (2013)
  • C. Madore et al.

    Early morphofunctional plasticity of microglia in response to acute lipopolysaccharide

    Brain Behav. Immun.

    (2013)
  • C. Madore et al.

    Nutritional n-3 PUFAs deficiency during perinatal periods alters brain innate immune system and neuronal plasticity-associated genes

    Brain Behav. Immun.

    (2014)
  • A. Moranis et al.

    Long term adequate n-3 polyunsaturated fatty acid diet protects from depressive-like behavior but not from working memory disruption and brain cytokine expression in aged mice

    Brain Behav. Immun.

    (2012)
  • M. Murthy et al.

    Differential effects of n-3 fatty acid deficiency on phospholipid molecular species composition in the rat hippocampus

    J. Lipid Res.

    (2002)
  • W.S. Powell et al.

    Biosynthesis, biological effects, and receptors of hydroxyeicosatetraenoic acids (HETEs) and oxoeicosatetraenoic acids (oxo-ETEs) derived from arachidonic acid

    BBA

    (2015)
  • D. Pratico et al.

    12/15-lipoxygenase is increased in Alzheimer's disease: possible involvement in brain oxidative stress

    Am. J. Pathol.

    (2004)
  • N. Quan et al.

    Cyclooxygenase 2 mRNA expression in rat brain after peripheral injection of lipopolysaccharide

    Brain Res.

    (1998)
  • C. Rey et al.

    Resolvin D1 and E1 promote resolution of inflammation in microglial cells in vitro

    Brain Behav. Immun.

    (2016)
  • C.N. Serhan et al.

    The resolution code of acute inflammation: novel pro-resolving lipid mediators in resolution

    Semin. Immunol.

    (2015)
  • L. Sun et al.

    12/15-Lipoxygenase metabolites of arachidonic acid activate PPARgamma: a possible neuroprotective effect in ischemic brain

    J. Lipid Res.

    (2015)
  • M.N. Woodroofe et al.

    Cytokine mRNA expression in inflammatory multiple sclerosis lesions: detection by non-radioactive in situ hybridization

    Cytokine

    (1993)
  • L. Yu et al.

    Time course change of COX2-PGI2/TXA2 following global cerebral ischemia reperfusion injury in rat hippocampus

    Behav. Brain Funct.

    (2014)
  • Y. Zhou et al.

    Soluble epoxide hydrolase inhibitor attenuates lipopolysaccharide-induced acute lung injury and improves survival in mice

    Shock

    (2017)
  • M. Antonietta Ajmone-Cat et al.

    Docosahexaenoic acid modulates inflammatory and antineurogenic functions of activated microglial cells

    J. Neurosci. Res.

    (2012)
  • M.G. Balvers et al.

    Fish oil and inflammatory status alter the n-3 to n-6 balance of the endocannabinoid and oxylipin metabolomes in mouse plasma and tissues

    Metabolomics

    (2012)
  • R.P. Bazinet et al.

    Polyunsaturated fatty acids and their metabolites in brain function and disease

    Nat. Rev. Neurosci.

    (2014)
  • M. Birnie et al.

    Temporal changes of cytochrome P450 (Cyp) and eicosanoid-related gene expression in the rat brain after traumatic brain injury

    BMC Genom.

    (2013)
  • V. Blais et al.

    Role of the innate immune response in the brain

    Med. Sci. (Paris)

    (2003)
  • P.K. Chang et al.

    Docosahexaenoic acid (DHA): a modulator of microglia activity and dendritic spine morphology

    J. Neuroinflam.

    (2015)
  • R. Chianese et al.

    Impact of dietary fats on brain functions

    Curr. Neuropharmacol.

    (2018)
  • D.W. Chung et al.

    Systemic administration of lipopolysaccharide induces cyclooxygenase-2 immunoreactivity in endothelium and increases microglia in the mouse hippocampus

    Cell. Mol. Neurobiol.

    (2010)
  • V. De Smedt-Peyrusse et al.

    Docosahexaenoic acid prevents lipopolysaccharide-induced cytokine production in microglial cells by inhibiting lipopolysaccharide receptor presentation but not its membrane subdomain localization

    J. Neurochem.

    (2008)
  • J.C. Delpech et al.

    Transgenic increase in n-3/n-6 fatty acid ratio protects against cognitive deficits induced by an immune challenge through decrease of neuroinflammation

    Neuropsychopharmacology

    (2015)
  • J.C. Delpech et al.

    Dietary n-3 PUFAs deficiency increases vulnerability to inflammation-induced spatial memory impairment

    Neuropsychopharmacology

    (2015)
  • F. Destaillats et al.

    Differential effect of maternal diet supplementation with alpha-Linolenic adcid or n-3 long-chain polyunsaturated fatty acids on glial cell phosphatidylethanolamine and phosphatidylserine fatty acid profile in neonate rat brains

    Nutr. Metab. (Lond.)

    (2010)
  • P. Dieter et al.

    LPS-induced synthesis and release of PGE2 in liver macrophages: regulation by CPLA2, COX-1, COX-2, and PGE2 synthase

    Adv. Exp. Med. Biol.

    (2002)
  • Cited by (0)

    View full text