Lipids

Makrides M, Ochoa JB, Szajewska H (eds): The Importance of Immunonutrition. Nestlé Nutr Inst Workshop Ser, vol 77, pp 121–131, (DOI: 10.1159/000351395) Nestec Ltd., Vevey/S. Karger AG., Basel, © 2013

Docosahexaenoic Acid and Its Derivative Neuroprotectin D1 Display Neuroprotective Properties in the Retina, Brain and Central Nervous System

Nicolas G. Bazan • Jorgelina M. Calandria • William C. Gordon Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, School of Medicine, New Orleans, LA , USA

Abstract The significance of the selective enrichment in omega-3 essential fatty acids (docosa- hexaenoyl – DHA – chains of membrane phospholipids, 22C and 6 double bonds) in the nervous system (e.g. synaptic membranes and dendrites) has remained, until recently, incompletely understood. While studying mechanisms of neuronal survival, we contrib- uted to the discovery of a docosanoid synthesized by 15--1 from DHA, which

we dubbed neuroprotectin D1 (NPD1; 10R,17S-dihydroxy-docosa-4Z,7Z,11E,13E,15E,19Z hexaenoic acid). NPD1 is a docosanoid because it is derived from a 22C precursor (DHA), unlike eicosanoids, which are derived from the 20C arachidonic acid family of essential fatty acids not enriched in the nervous system. We found that NPD1 is promptly made in response to oxidative stress, seizures and brain ischemia-reperfusion. NPD1 is neuropro- tective in experimental brain damage, retinal pigment epithelial cells, and in human brain cells. Thus, NPD1 acts as a protective sentinel, one of the very first defenses activated when cell homeostasis is threatened by neurodegenerations. NPD1 also has been shown to have a specificity and potency that provides beneficial bioactivity during initiation and early progression of neuronal and retinal degenerations, epilepsy and stroke. In short, NPD1 regulation promotes homeostatic regulation of neural circuitry integrity. Copyright © 2013 Nestec Ltd., Vevey/S. Karger AG, Basel Introduction

The interface between the immune and inflammatory systems actively influ- ences the development and maintenance of the central nervous system (CNS) by sustaining homeostasis. This interface is critical to preserve the cellular and functional integrity of the nervous system during aging as well. Moreover, most diseases and dysfunctions of the nervous system display failures in early re- sponses from the immune and inflammatory systems. The selective uptake and avid retention of the omega-3 essential fatty acid (FA; fig. 1 ) (DHA) by the CNS is a salient characteristic necessary for synaptogenesis as well as development and biogenesis of all neu- ronal cellular membranes. This also includes photoreceptors since the retina is an integral part of the CNS. Unlike members of the omega-6 FAs (e.g. arachi- donic acid), which are present in all parts of the body, DHA is selectively taken up by the CNS and is needed for a variety of functions, including memory and vision. DHA is concentrated and retained in the nervous system and becomes part of the membranes. DHA is also used in development of cellular elements that comprise the neurovascular unit, the innate immune resident cells, the mi- croglia, and the astrocytes. During the last few years, bioactive messengers derived from DHA have been identified, and these discoveries have opened up a renewed interest in under- standing the bioactivity and significance of this essential FA in the nervous s ystem. The first bio-derivative of DHA, neuroprotectin D1 (NPD1; 10R,17S-dihy- droxy-docosa-4Z,7Z,11E,13E,15E,19Z hexaenoic acid) was found to be made on-demand in the CNS. NPD1 biosynthesis is activated at the onset of various injuries, including ischemia-reperfusion damage, protein misfolding, Aβ pep- tide challenge, enhanced oxidative stress, and others (fig. 1 ). This stereospe- cific mediator is secreted upon synthesis, extending its neuroprotective prop- erties to the vicinity of the injury. These characteristics, along with the pro- homeostatic form of action that NPD1 elicits, make it a useful and interesting molecule for the development of new therapies to treat neurodegenerative diseases.

Docosahexaenoic Acid and Docosanoids

Docosanoids are derived from 22C DHA and include NPD1, [1] , neu- roprostanes, and related 22C derivatives, unlike eicosanoids, which are derived from the 20C arachidonic acid family of essential FAs ( fig. 1 ). Previous studies

122 Bazan · Calandria · Gordon Eicosanoids and docosanoids

FA FA Free AA PLA2 20:4 22:6 PLA2 Free DHA P cholineP choline COX-1,-2 15-LO-1 Eicosanoids Neurotrophins Docosanoid (e.g. prostaglandins, (e.g. PEDF, BDNF, (NPD1) prostacyclin, thromboxanes, CNTF, LIF, NT3 leukotrienes, lipoxins (Sterospecific F-4 neuroprostane binding site) Receptors Free radical-mediated FA oxidation Bioactivity Bioactivity HNE HHE

‡ PPR-Dž ‡ Akt-1 (Akt/PKB) Proinflammatory ‡ Cornea nerve regeneration Gene expression Antiapoptotic ‡ Antiangiogenic ‡ COX-2 ‡ Proapoptotic Bcl-2 proteins ‡ Protein misfolding ‡ CEX-1 Bcl-2 proteins ‡ PP2A-BCLXL-Bax ‡ Transcriptional regulation ‡ B-94 ‡ ARE

Fig. 1. Omega-3 and omega-6 FA mechanisms of cell death and survival. Phospholipids carrying both omega-3 and omega-6 FAs form part of the cellular membrane. DHA with a chain of 22 carbons is avidly retained in cell membranes of the CNS as opposed to

arachidonic acid (AA; 20 carbons) that occurs ubiquitously. Phospholipase A2 (PLA2 ) hy- drolyzes both FA types from the second position in the phospholipids of the plasma membrane, and they are converted into bioactive signaling lipids. AA is the precursor of prostaglandins, prostacyclins, thromboxanes, and leukotrienes, among others. Cyclo- oxygenase-1 (COX-1) and COX-2 are the main enzymes that convert AA into their bioac- tive lipids. These immunologically related messengers form part of the signaling, which mediates cellular response and , acting through specific receptors to elicit their bioactivity. Peroxidation products of AA and DHA produce 4-hydroxynonenal (4- HNE) and 4-hydroxyhexenal (4-HHE), which are toxic byproducts of generalized oxida- tive stress. On the other hand, 15-LOX-1 is the enzyme that converts DHA into its stereo- specific dioxygenation product NPD1. NPD1 has stereospecific binding that triggers modulation of proinflammatory and proapoptotic processes by altering the expression and phosphorylation state of Bcl-2 proteins COX-2, CEX-1 and B-94. It also enhances the activation of pro-survival key kinases such as Akt. Altogether, the activity of NPD1 pro- motes corneal nerve regeneration as well as survival of neuronal and other retinal cells. The synthesis of NPD1 is greatly enhanced by the action of neurotrophins, including PEDF and BDNF.

Neuroprotective Properties of DHA and NPD1 123 identified the formation of mono-, di-, and trihydroxy DHA derivatives in the retina. These studies show that lipoxygenase inhibitors block synthesis of these derivatives, indicating an enzymatic process of a lipoxygenase nature [2] . Al- though the stereochemistry and bioactivity of DHA-oxygenated derivatives were not determined at that time, it was proposed that these DHA derivatives could be neuroprotective, and thus the term ‘docosanoids’ was coined [3, 4] . Liquid chromatography-photodiode array-electrospray ionization tandem mass spectrometry-based lipidomic analysis was then used to identify oxygenation pathways for synthesis of NPD1 during brain ischemia-reperfusion [5], in epi- lepsy, in the retinal pigment epithelium (RPE) [6] , in the human brain [7] , and in other cells [8, 9] .

Docosahexaenoic Acid and Neuroprotectin D1

The stereospecific isomer NPD1 is derived from the enzymatic dioxygenation of DHA (fig. 1 ). Photoreceptors, RPE cells, and the brain contain unesterified (free) DHA under basal, nonstimulated conditions [10–14] , suggesting that the pool size of unesterified DHA is tightly regulated by phospholipase A 2 , reacyla- tion, and peroxidation under some conditions. Free DHA becomes the substrate of docosahexaenoyl-coenzyme A synthesis for channeling through acyltransfer- ases, which incorporate this FA into phospholipids [15–18]. RPE cells therefore modulate the uptake, conservation, and delivery of DHA to the photoreceptors [4] . In addition, RPE cells use a specific pool of DHA phospholipids to initiate NPD1 synthesis by 15-lipoxygenase-1 (15-LOX-1) [19] ( fig. 1 ). NPD1 induces homeostatic/pro-survival signaling on cells undergoing stress as a result of cellular and systemic damage [3, 5, 7] . For instance, NPD1 induces in brain ischemia-reperfusion and in oxidative- stressed retinal cells [5, 20, 21] and inhibits retinal ganglion cell death [21] , is protective in kidney ischemia-reperfusion and regulates adiponectin [22] . In addition, NPD1 upregulates antiapoptotic proteins (Bcl-2 and Bcl-xL) and downregulates proapoptotic proteins (Bax and Bad) in response to cellular ox- idative stress and cytokine activation leading to an overall pro-survival tran- scriptome [3, 5–7] . NPD1 also provides the basis to understand the mechanisms by which DHA promotes survival during neuroinflammation and neurodegeneration as it is the mediator that executes protective bioactivity of DHA in the CNS. Deficiency of NPD1 and 15-LOX-1, the enzyme involved in its formation, has been observed in Alzheimer’s disease brain [7]. Also, NPD1 further influences β-amyloid pre- cursor protein processing and decreases amyloid-β (Aβ) 42 release [23] . DHA,

124 Bazan · Calandria · Gordon the precursor of NPD1, elicits an Aβ 42 -lowering effect both in vitro and in vivo [24–26] . Conversely, DHA effects differ from NPD1-triggered ones in some instances. For example, it is worthy to note that free radical-mediated DHA peroxidation products accumulate during ischemia and neurodegeneration (fig. 1 ). These ox- idation products in turn may form protein adducts and other cytotoxic mole- cules that promote further free radical injury [27–29]. Enzymatic dioxygenation of DHA, as opposed to peroxidation, produces stereospecific products, such as NPD1, that are controlled, produced and secreted [19] . NPD1 also stereo-selec- tively modulates pathways triggered by insults such as oxidative stress in femto- molar doses [19]. Thus, NPD1 activity may explain at least in part the pro-sur- vival properties of DHA.

Docosahexaenoic Acid and Neuroprotectin D1 in Photoreceptor Cell Membranes

Photoreceptor cell damage and apoptotic death are tell-tale signs of retinal de- generative diseases [30–32] . In retinitis pigmentosa (RP), rod photoreceptor death begins on the edges of the retina, while in age-related macular degenera- tion (AMD) progressive agitation and vision loss result from photoreceptor death in the macula, the center of the retina [33] . Retinal degenerations include several cell-signaling pathways, such as various triggers of photoreceptor cell death. This death is closely linked with the relationship between photoreceptors and RPE cells. For example, photoreceptors are damaged in diseases such as Stargardt’s disease and other retinal degenerations where RPE cell integrity is compromised. If RPE cells die, photoreceptor cells also succumb [34] . There are more than 150 mutations of photoreceptor-specific proteins reported to con- tribute to RP, including mutations of rhodopsin, peripherin, the β-subunit of cGMP phosphodiesterase, and photoreceptor outer segment membrane protein 1 (ROM1) [35, 36] . In autosomal-dominant RP, some of the genes involved are expressed in the choriocapillaris [37]. AMD, however, is a group of complex disorders with multiple environmental and genetic causes. The main known risk factor is advanced age. Moreover, the choriocapillaris and Bruch’s membrane, which separates the RPE from the choriocapillaris, also are affected in AMD [38] . Initiation and progression of this retinal degeneration involves an unsuc- cessful inflammatory response. Recent findings have identified major risk fac- tors for AMD: single nucleotide polymorphisms in the genes encoding factor H (CFH/HF1) [39], factor B (BF), and complement component 2 (C2) [40] . Factor H is an inhibitor of the alternative pathway of complement system activation

Neuroprotective Properties of DHA and NPD1 125 that limits cell injury and inflammation [41]. Nevertheless, our knowledge on the mechanisms leading to these diseases is limited. Complex intracellular and intercellular signaling events are set in motion during the preliminary stages of retinal degeneration. During these stages, oxi- dative and nitrosative stress is increased and exaggerated, usually at abnormal cellular locations and with inappropriate timing. Mitochondrial function is also compromised, and this dysfunction is a major factor in these impediments [37, 42, 43] . NPD1 has also been shown to inhibit choroidal neovascularization (CNV) in a laser-induced mouse model of CNV [44] . This model shows how NPD1 protects photoreceptor cells from apoptosis while downregulating the proin- flammatory cytokines TNF-α and IL-1β, possibly by inhibiting NF-κB activity and reducing cyclooxygenase-2, leading to decreased VEGF expression. Overall, NPD1 may contribute to induction of the resolution phase of the inflammatory response ( fig. 1 ). The antiangiogenic and neuroprotective actions of NPD1 make it an excellent candidate for treating wet AMD and diabetic retinopathy. Because early clinical expressions of most retinal degenerations precede pho- toreceptor cell death, it is important to determine these early critical events. This knowledge may lead to new therapeutic interventions for halting or slowing dis- ease progression.

Docosahexaenoic Acid and Neuroprotectin D1 in Stroke

Stroke is the third leading cause of death and the first cause of adult disability in the United States. In spite of the knowledge gained regarding stoke pathophysi- ology, the only treatment approved for ischemic stroke is thrombolysis, and only 3–5% of patients can undergo this treatment. Thus, there is a critical need for developing effective stroke treatments. Cerebral ischemia results in rapid accumulation of free FAs, such as DHA released from membrane phospholipids. DHA is concentrated in phospholipids of various cells of the CNS [11] and is processed in the liver for transport to various tissues by the bloodstream. DHA release from cell membranes mostly occurs under conditions of excessive oxidative stress [11]. As an acyl chain of membrane phospholipids, DHA is necessary for ion channels, receptors, and transporters to maintain proper physical properties [11]. It is involved in mem- ory formation [45] , excitable membrane function [46] , and neuroprotection [47, 48] . As mentioned previously, DHA is the precursor of NPD1, which downregu- lates apoptosis and promotes cell survival [6, 49, 50]. In addition, some studies

126 Bazan · Calandria · Gordon show that NPD1 inhibits brain ischemia-reperfusion-mediated leukocyte infil- tration and proinflammatory gene expression [49]. It also promotes in vitro and in vivo neurogenesis [5] (fig. 1 ). NPD1 displays potent anti-inflammatory ac- tions and protects against brain ischemia-reperfusion [5] and oxidative stress [3, 6, 7] . Recent compelling evidence gathered in our lab shows the neuroprotective properties of DHA when it is systemically administered in a rat model of mid- dle cerebral artery occlusion (MCAo). DHA improves neurological function in rats and reduces the extent of histological damage with a therapeutic win- dow of at least 3 h after MCAo onset. As such, DHA offers great promise in developing therapies for cerebral ischemia in patients with acute ischemic stroke. Again, one of the possible mechanisms of neuroprotection mediated by DHA may involve its conversion into bioactive lipid mediators such as NPD1.

Docosahexaenoic Acid and Neuroprotectin D1 in Epilepsy

Temporal lobe epilepsy is a disorder where hippocampal function is disrupted by neuronal hyperexcitability [51–53] , often spreading from the hippocampus to other areas of the brain [54] . Under resting conditions, unesterified DHA is present in the brain in small amounts. However, when seizures induce the activation of phospholipases, es- terified DHA is released and it accumulates in hippocampal synaptosomes [55] . DHA has been proposed to elicit anti-seizure activity through modulation of ion channels and neurotransmitter receptors, as well as regulation of synaptic plas- ticity [56–59] . Progressive and permanent enhancement of local and propagating seizure activity occurs in response to repeated hippocampal stimulation in a kindling epileptogenesis model for temporal lobe epilepsy [60–63] . Here, DHA and NPD1 modulate the hippocampal electrical activity associated with evoked sei- zures, thereby limiting the progression of focal limbic seizures. Also, DHA sig- nificantly increases seizure latency induced by pentylenetetrazol (PTZ) [64] . The evidence suggests that NPD1 may attenuate the onset of PTZ-induced sei- zures and ameliorate enhanced electrical activity. In this promising field, future studies are required to determine the effect of oral and systemic DHA adminis- tration on seizure frequency in different experimental epilepsy models for po- tential translational research. As such, both DHA and NPD1 make attractive targets in the development of therapeutic approaches for treating and prevent- ing epilepsy.

Neuroprotective Properties of DHA and NPD1 127 Conclusions

Omega-3 FAs are essential for the body, and supplementation may have impor- tant implications for human health, particularly in the CNS. Dietary supplemen- tation may positively influence dyslipidemia, atherosclerosis, hypertension, dia- betes mellitus, obesity, metabolic syndrome, and other inflammatory diseases. Fish, fish oils, and some vegetable oils are rich sources of omega-3 FAs. On the other hand, a recent prospective analysis does not support the concept that long- term dietary intake of omega-3 FAs decreases the risk of type 2 diabetes. In con- trast, higher fish and omega-3 FA consumption appears to be associated with higher incidence of type 2 diabetes. Given the beneficial effects of fish and ome- ga-3 FAs on multiple risk factors associated with diabetes, including triglycer- ides, high-density lipoprotein cholesterol, blood pressure, and inflammation, and on coronary heart disease, the major sequela of diabetes, the clinical rele- vance of this relation and its possible mechanisms require further investigation. Ultimately, clinical trials using various DHA formulations will need to dem- onstrate the safety and efficacy of these interventions. Because omega-3 FAs (mainly DHA) are liable to peroxidation, DHA signalolipidomics research on the molecular details of DHA uptake, disposition, and action will likely identify other factors/molecules/systems that protect DHA integrity, particularly during the initiation and early progression of diseases. These new approaches may lead to novel multinutrient-based trials in the future. Both DHA and its stereospecific derivative NPD1 have been shown to have neuroprotective roles in the retina and CNS. Future studies are needed to help garner information for developing therapies to conquer diseases such as RP, AMD, stroke, epilepsy, Alzheimer’s and other neural and retinal degenerative diseases.

Acknowledgements

This work was supported by grants from the National Institutes of Health, National In- stitute of General Medical Sciences (GM103340), National Eye Institute (EY005121) and the National Institute of Neurological Disorders and Stroke (NS046741).

Disclosure Statement

The authors have no conflicts of interest to disclose.

128 Bazan · Calandria · Gordon References

1 Serhan CN, Hong S, Gronert K, et al: Re- 11 Aveldano MI, Bazan NG: Differential lipid solvins: a family of bioactive products of deacylation during brain ischemia in a omega-3 fatty acid transformation circuits homeotherm and a poikilotherm. Content initiated by aspirin treatment that counter and composition of free fatty acids and triac-

proinflammation signals. J Exp Med 2002; ylglycerols. Brain Res 1975; 100: 99–110.

196: 1025–1037. 12 Bazan NG: Synaptic : signifi- 2 Bazan NG, Birkle DL, Reddy TS: Docosa- cance of polyunsaturated fatty acids and

hexaenoic acid (22: 6, n-3) is metabolized to platelet-activating factor. J Lipid Res 2003; 44: lipoxygenase reaction products in the retina. 2221–2233.

Biochem Biophys Res Commun 1984; 125: 13 Horrocks LA, Farooqui AA: NMDA recep- 741–747. tor-stimulated release of arachidonic acid: 3 Bazan NG: Homeostatic regulation of photo- mechanisms for the Bazan effect; in Municio receptor cell integrity: significance of the po- AM, Miras-Portugal MT (eds): Cell Signal tent mediator neuroprotectin D1 biosynthe- Transduction, Second Messengers, and Pro- sized from docosahexaenoic acid: the Proctor tein Phosphorylation in Health and Disease.

Lecture. Invest Ophthalmol Vis Sci 2007; 48: New York, Plenum, 1994, pp 113–128. 4866–4881. 14 Sun GY, Xu J, Jensen MD, Simonyi A: Phos- 4 Bazan NG, Birkle DL, Reddy TS: Biochemical pholipase A2 in the central nervous system: and nutritional aspects of the metabolism of implications for neurodegenerative diseases. J

polyunsaturated fatty acids and phospholip- Lipid Res 2004; 45: 205–213. ids in experimental models of retinal degen- 15 Reddy TS, Bazan NG: Activation of polyun- eration; in LaVail MM, Hollyfield JG, Ander- saturated fatty acids by rat tissues in vitro.

son RE (eds): Retinal Degeneration: Lipids 1984; 19: 987–989. Experimental and Clinical Studies. New 16 Reddy TS, Bazan NG: Synthesis of arachi- York, Liss, 1985, pp 159–187. donoyl coenzyme A and docosahexaenoyl

5 Marcheselli VL, Hong S, Lukiw WJ, et al: coenzyme A in retina. Curr Eye Res 1984; 3: Novel docosanoids inhibit brain ischemia- 1225–1232. reperfusion-mediated leukocyte infiltration 17 Reddy TS, Bazan NG: Synthesis of arachi- and pro-inflammatory gene expression. J Biol donoyl coenzyme A and docosahexaenoyl

Chem 2003; 278: 43807–43817. coenzyme A in synaptic plasma membranes 6 Mukherjee PK, Marcheselli VL, Serhan CN, of cerebrum and microsomes of cerebrum, Bazan NG: Neuroprotectin D1: a docosa- cerebellum, and brain stem of rat brain.

hexaenoic acid-derived docosatriene protects J Neurosci Res 1985; 13: 381–390. human retinal pigment epithelial cells from 18 Reddy TS, Bazan NG: Synthesis of docosa- oxidative stress. Proc Natl Acad Sci USA hexaenoyl-, arachidonoyl- and palmitoyl-co-

2004; 101: 8491–8496. enzyme A in ocular tissues. Exp Eye Res

7 Lukiw WJ, Cui J, Marcheselli VL, et al: A role 1985b;41: 87–95. for docosahexaenoic acid-derived neuropro- 19 Calandria JM, Marcheselli VL, Mukherjee tectin D1 in neural cell survival and Alzheim- PK, et al: Selective survival rescue in 15-li-

er disease. J Clin Invest 2005; 115: 2774–2783. poxygenase-1-deficient retinal pigment epi- 8 Schwab JM, Chiang N, Arita M, Serhan CN: thelial cells by the novel docosahexaenoic E1 and protectin D1 activate inflam- acid-derived mediator, neuroprotectin D1.

mation-resolution programmes. Nature 2007; J Biol Chem 2009; 284: 17877–17882.

447: 869–874. 20 Mukherjee PK, Marcheselli VL, de Rivero 9 Serhan CN: Resolution phase of inflamma- Vaccari JC, et al: Photoreceptor outer seg- tion: novel endogenous anti-inflammatory ment phagocytosis attenuates oxidative and proresolving lipid mediators and path- stress-induced apoptosis with concomitant

ways. Annu Rev Immunol 2007; 25: 101–137. neuroprotectin D1 synthesis. Proc Natl Acad

10 Aveldano MI, Bazan NG: Displacement into Sci USA 2007; 104: 13158–13163. incubation medium by albumin of highly un- saturated retina free fatty acids arising from

membrane lipids. FEBS Lett 1974; 40: 53–56.

Neuroprotective Properties of DHA and NPD1 129 21 Qin Q, Patil KA, Gronert K, Sharma SC: 32 Papermaster DS: The birth and death of pho- Neuroprotectin D1 inhibits retinal ganglion toreceptors: the Friedenwald Lecture. Invest

cell death following axotomy. Prostaglandins Ophthalmol Vis Sci 2002; 43: 1300–1309.

Leukot Essent Fatty Acids 2008; 79: 201–207. 33 Bird AC: The Bowman lecture. Towards an 22 González-Périz A, Horrillo R, Ferré N, et al: understanding of age-related macular disease.

Obesity-induced insulin resistance and he- Eye 2003; 17: 457–466. patic steatosis are alleviated by omega-3 fatty 34 Cideciyan AV, Aleman TS, Swider M, et al: acids: a role for and protectins. Mutations in ABCA4 result in accumulation

FASEB J 2009; 23: 1946–1957. of lipofucsin before slowing the retinoid cy- 23 Zhao Y, Calon F, Julien C, et al: Docosa- cle: a reappraisal of the human disease se-

hexaenoic acid-derived neuroprotectin D1 quence. Hum Mol Genet 2004; 13: 525–534. induces neuronal survival via secretase- and 35 Dryja TP, McGee TL, Reichel E, et al: A point PPARγ-mediated mechanisms in Alzheimer’s mutation of the rhodopsin gene in one form

disease models. PLoS One 2011; 6:e15816. of retinitis pigmentosa. Nature 1990; 343: 24 Lim GP, Calon F, Morihara T, et al: A diet 364–366. enriched with the omega-3 fatty acid docosa- 36 Liu Q, Zuo J, Pierce EA: The retinitis pig- hexaenoic acid reduces amyloid burden in an mentosa 1 protein is a photoreceptor micro-

aged Alzheimer mouse model. J Neurosci tubule-associated protein. J Neurosci 2004;

2005; 25: 3032–3040. 24: 6427–6436. 25 Oksman M, Iivonen H, Hogyes E, et al: Im- 37 Mendes HF, van der Spuy J, Chapple JP, pact of different saturated fatty acid, polyun- Cheetham ME: Mechanisms of cell death in saturated fatty acid and cholesterol contain- rhodopsin retinitis pigmentosa: implications

ing diets on beta-amyloid accumulation in for therapy. Trends Mol Med 2005; 11: 177– APP/PS1 transgenic mice. Neurobiol Dis 185.

2006; 23: 563–572. 38 Chong NH, Keonin J, Luthert PJ, et al: De- 26 Sahlin C, Pettersson FE, Nilsson LN, et al: crease thickness and integrity of the macular Docosahexaenoic acid stimulates non-amy- elastic layer of Bruch’s membrane correspond loidogenic APP processing resulting in re- to the distribution of lesions associated with duced Abeta levels in cellular models of Al- age-related macular degeneration. Am J

zheimer’s disease. Eur J Neurosci 2007; 26: Pathol 2005; 166: 241–251. 882–889. 39 Hageman GS, Anderson DH, Johnson LV, et 27 Galasko D, Montine TJ: Biomarkers of oxida- al: A common haplotype in the complement tive damage and inflammation in Alzheimer’s regulatory gene factor H (HF1/CFH) predis-

disease. Biomark Med 2010; 4: 27–436. poses individuals to age-related macular de-

28 Musiek ES, Brooks JD, Joo M, et al: Electro- generation. Proc Natl Acad Sci USA 2005;

philic cyclopentenone neuroprostanes are 102: 7227–7232. anti-inflammatory mediators formed from 40 Gold B, Merriam JE, Zernant J, et al: Varia- the peroxidation of the omega-3 polyunsatu- tion in factor B (BF) and complement 2 (C2) rated fatty acid docosahexaenoic acid. J Biol genes is associated with age-related macular

Chem 2008; 283: 19927–19935. degeneration. Nat Genet 2006; 38: 458–462. 29 Roberts LJ, Fessel JP, Davies SS: The bio- 41 Bok D: Evidence for an inflammatory process chemistry of the isoprostane, neuroprostane, in age-related macular degeneration gains

and isofuran pathways of lipid peroxidation. new support. Proc Natl Acad Sci USA 2005;

Brain Pathol 2005; 15: 143–148. 102: 7053–7054. 30 Chang GQ, Hao Y, Wong F: Apoptosis: final 42 Sreekumar PG, Kannan R, Yaung J, et al: Pro- common pathway of photoreceptor death in tection from oxidative stress by methionine rd, rds, and rhodopsin mutant mice. Neuron sulfoxide reductases in RPE cells. Biochem

1993; 11: 595–605. Biophys Res Commun 2005; 334: 245–253. 31 Portera-Cailliau C, Sung CH, Nathans J, 43 Feher J, Kovacs I, Artico M, et al: Mitochon- Adler R: Apoptotic photoreceptor cell death drial alterations of retinal pigment epithelium in mouse models of retinitis pigmentosa. in age-related macular degeneration. Neuro-

Proc Natl Acad Sci USA 1994; 91: 974–978. biol Aging 2006; 27: 983–993.

130 Bazan · Calandria · Gordon 44 Sheets KG, Zhou Y, Ertel MK, et al: Neuro- 55 Birkle DL, Bazan NG: Effect of bicuculline- protectin D1 attenuates laser-induced choroi- induced status epilepticus on prostaglandins dal neovascularization in mouse. Mol Vis and hydroxyeicosatetraenoic acids in rat

2010; 16: 320–329. brain subcellular fractions. J Neurochem

45 Gamoh S, Hashimoto M, Sugioka K, et al: 1987; 48: 1768–1778. Chronic administration of docosahexaenoic 56 Champeil-Potokar G, Chaumontet C,

acid improves reference memory-related Guesnet P, et al: Docosahexaenoic acid (22: learning ability in young rats. Neuroscience 6n-3) enrichment of membrane phospholip-

1999; 93: 237–241. ids increases gap junction coupling capacity

46 McGahon BM, Martin DS, Horrobin DF, in cultured astrocytes. Eur J Neurosci 2006;

Lynch MA: Age-related changes in synaptic 24: 3084–3090. function: analysis of the effect of dietary sup- 57 Poling JS, Vicini S, Rogawski MA, Salem N plementation with omega-3 fatty acids. Neu- Jr: Docosahexaenoic acid block of neuronal + roscience 1999; 94: 305–314. voltage-gated K channels: subunit selective 47 Rodriguez de Turco EB, Belayev L, Liu Y, et antagonism by zinc. Neuropharmacology

al: Systemic fatty acid responses to transient 1996; 35: 969–982. focal cerebral ischemia: influence of neuro- 58 Xiao Y, Li X: Polyunsaturated fatty acids protectant therapy with human albumin. J modify mouse hippocampal neuronal excit-

Neurochem 2002; 83: 515–524. ability during excitotoxic or convulsant stim-

48 Belayev L, Marcheselli VL, Khoutorova L, et ulation. Brain Res 1999; 846: 112–121. al: Docosahexaenoic acid complexed to albu- 59 Young C, Gean PW, Wu SP, et al: Cancella- min elicits high-grade ischemic neuroprotec- tion of low-frequency stimulation-induced

tion. Stroke 2005; 36: 118–123. long-term depression by docosahexaenoic 49 Serhan CN, Gotlinger K, Hong S, et al: Anti- acid in the rat hippocampus. Neurosci Lett

inflammatory actions of neuroprotectin D1/ 1998; 247: 198–200. protectin D1 and its natural stereoisomers: 60 Young C, Gean PW, Chiou LC, Shen YZ: assignments of dihydroxy-containing docosa- Docosahexaenoic acid inhibits synaptic

trienes. J Immunol 2006; 176: 1848–1859. transmission and epileptiform activity in the

50 Mukherjee PK, Chawla A, Loayza MS, Bazan rat hippocampus. Synapse 2000; 37: 90–94. NG: Docosanoids are multifunctional regula- 61 Cole-Edwards KK, Musto AE, Bazan NG: tors of neural cell integrity and fate: signifi- c-Jun N-terminal kinase activation responses cance in aging and disease. Prostaglandins induced by hippocampal kindling are medi-

Leukot Essent Fatty Acids 2007; 77: 233–238. ated by reactive astrocytes. J Neurosci 2006;

51 Masukawa LM, Higashima M, Kim JH, Spen- 26: 8295–8304. cer DD: Epileptiform discharges evoked in 62 Morimoto K, Fahnestock M, Racine RJ: Kin- hippocampal brain slices from epileptic pa- dling and status epilepticus models of epilep-

tients. Brain Res 1989; 493: 168–174. sy: rewiring the brain. Prog Neurobiol 2004;

52 Schwartzkroin PA: Hippocampal slices in 73: 1–60. experimental and human epilepsy. Adv Neu- 63 Sutula T, Koch J, Golarai G, et al: NMDA re-

rol 1986; 44: 991–1010. ceptor dependence of kindling and mossy 53 Williamson A, Patrylo PR, Pan J, et al: Corre- fiber sprouting: evidence that the NMDA re- lations between granule cell physiology and ceptor regulates patterning of hippocampal

bioenergetics in human temporal lobe epilep- circuits in the adult brain. J Neurosci 1996;

sy. Brain 2005; 128: 1199–1208. 16: 7398–7406. 54 Umeoka S, Terada K, Baba K, et al: Neural 64 Taha AY, Bunham WM, Auvin S: Polyunsat-

connection between bilateral basal temporal urated fatty acid and epilepsy. Epilepsia 2010;

regions: cortico-cortical evoked potential 5: 1348–1358. analysis in patients with temporal lobe epi-

lepsy. Neurosurgery 2009; 64: 847–855.

Neuroprotective Properties of DHA and NPD1 131