Omega-3 Research papers – Reference
No human clinical trial data are reported for supplementation benefits of omega-3 fatty acids in patients with either the traumatic brain injury or ischemic reperfusion injury. Although supplementation benefits of omega-3 fatty acids in patients with brain injuries have not been reported, data from several preclinical trials suggest that either pre- or post-supplementation of omega-3 fatty acids could benefit patients with brain injuries. The following is a list of relevant papers from NIH’s PubMed:
1. Lipids Health Dis. 2013 Feb 11;12(1):16. [Epub ahead of print] Modulating the inflammatory properties of activated microglia with Docosahexaenoic acid and Aspirin. Pettit LK, Varsanyi C, Tadros J, Vassiliou E. ABSTRACT: BACKGROUND: Microglia are considered the "resident macrophages" of the brain. When in their resting state, microglia perform routine maintenance and immune surveillance. Once activated, either by injury or an immune stimulus, microglia secrete a variety of pro-inflammatory molecules, such as Nitric Oxide, superoxide, and inflammatory cytokines. Up-regulation of pro-inflammatory molecules is transient, and does not cause neurodegeneration. However, if up-regulation lasts for an extended period of time, neurodegeneration ensues.Many neurodegenerative diseases are characterized by chronic inflammation due to microglial activation. Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) have been proposed as possible preventative treatments for neurodegenerative diseases, due to their anti-inflammatory properties. Docosahexaenoic Acid (DHA) is an omega-3 polyunsaturated fatty acid (PUFA) that has potent anti-inflammatory properties.This research work sought to elucidate whether microglial activation can be modulated by combining Aspirin, a classical NSAID, with Docosahexaenoic Acid, a natural anti-inflammatory agent. The combined ability of Aspirin and DHA to modulate microglial activation was determined in the context of pro-inflammatory cytokines, Nitric Oxide levels, as well as total Glutathione levels. RESULTS: Docosahexaenoic Acid increased total Glutathione levels in microglia cells and enhanced their anti-oxidative capacity. It reduced production of the pro-inflammatory cytokines TNF-alpha and IL-6 induced through TLR-3 and TLR-4 activation. Furthermore, it reduced production of Nitric Oxide. Aspirin showed similar anti-inflammatory effects with respect to TNF-alpha during TLR-3 and TLR-7 stimulation. Aspirin did not show any redection in terms of Nitric Oxide production. Combination of Aspirin and Docosahexaenoic Acid showed augmentation in total Glutathione production during TLR-7 stimulation as well as a reduction in IL-6, TNF-alpha and Nitric Oxide. CONCLUSIONS: Collectively, these findings highlight the combination of Docosahexaenoic Acid and Aspirin as a possible measure against inflammation of the nervous system, thus leading to protection against neurodegenerative diseases with an inflammatory etiology. PMID: 23398903 [PubMed - as supplied by publisher] 2. J Neurotrauma. 2013 Jan 30. [Epub ahead of print] OMEGA-3 FATTY ACIDS FOR TREATMENT OF TRAUMATIC BRAIN INJURY. Hasadsri L, Wang BH, Lee JV, Erdman JW, Llano DA, Barbey AK, Wszalek T, Sharrock MF, Wang H. Mayo Clinic, Laboratory Medicine and Pathology, Rochester, Minnesota, United States; hasadsri@life.illinois.edu. Traumatic Brain Injury (TBI) is a global public health epidemic. In the US alone, more than 3 million people sustain a TBI annually. It is one of the most disabling injuries as it results in motor and sensory deficits as well as severe cognitive, emotional, and psychosocial impairment, crippling many vital areas of higher functioning. Fueled by the recognition of TBI as the "signature injury" in our wounded soldiers in Iraq and Afghanistan and its often devastating impact on athletes playing contact sports, interest in TBI and TBI research has increased dramatically. Unfortunately, despite increased awareness of its detrimental consequences, there has been little progress in developing effective TBI interventions. Recent evidence, however, strongly indicates that nutritional intervention may provide a unique opportunity to enhance the neuronal repair process after TBI. To date, two omega-3 fatty acids, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), have the most promising laboratory evidence for their neuro-restorative capacities in TBI. Although both animal models and human studies of brain injuries suggest they may provide benefits, there has been no human trial evaluating the effects of n-3 fatty acids on resilience to, or treatment of, TBI. This paper reviews the known functions of n-3 fatty acids in the brain and their specific role in the cellular and biochemical pathways underlying neurotraumatic injury. We also highlight recent studies on the therapeutic impact of enhanced omega 3 intake in vivo, and how this may be a particularly promising approach to improving functional outcome in patients with TBI. PMID: 23363551 [PubMed - as supplied by publisher] 3. Am J Emerg Med. 2013 Jan;31(1):273.e5-8. doi: 10.1016/j.ajem.2012.05.014. Epub 2012 Aug 3. Therapeutic use of omega-3 fatty acids in severe head trauma. Lewis M, Ghassemi P, Hibbeln J. Brain Health Education and Research Foundation, Arlington, VA, USA. dr.michael.lewis@gmail.com Traumatic brain injury (TBI) has long been recognized as the leading cause of traumatic death and disability. Tremendous advances in surgical and intensive care unit management of the primary injury, including maintaining adequate oxygenation, controlling intracranial pressure, and ensuring proper cerebral perfusion pressure, have resulted in reduced mortality. However, the secondary injury phase of TBI is a prolonged pathogenic process characterized by neuroinflammation, excitatory amino acids, free radicals, and ion imbalance. There are no approved therapies to directly address these underlying processes. Here, we present a case that was intentionally treated with substantial amounts of omega-3 fatty acids (n-3FA) to provide the nutritional foundation for the brain to begin the healing process following severe TBI. Recent animal research supports the use of n-3FA, and clinical experience suggests that benefits may be possible from substantially and aggressively adding n-3FA to optimize the nutritional foundation of severe TBI patients and must be in place if the brain is to be given the opportunity to repair itself to the best possible extent. Administration early in the course of treatment, in the emergency department or sooner, has the potential to improve outcomes from this potentially devastating public health problem. Copyright © 2013 Elsevier Inc. All rights reserved. PMCID: PMC3518659 PMID: 22867826 [PubMed - in process] 4. PLoS One. 2013;8(1):e51543. doi: 10.1371/journal.pone.0051543. Epub 2013 Jan 4. Low vitamin D status and suicide: a case-control study of active duty military service members. Umhau JC, George DT, Heaney RP, Lewis MD, Ursano RJ, Heilig M, Hibbeln JR, Schwandt ML. National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA. umhau@jhu.edu OBJECTIVE: Considering that epidemiological studies show that suicide rates in many countries are highest in the spring when vitamin D status is lowest, and that low vitamin D status can affect brain function, we sought to evaluate if a low level of 25-hydroxyvitamin D [25(OH)D] could be a predisposing factor for suicide. METHOD: We conducted a prospective, nested, case-control study using serum samples stored in the Department of Defense Serum Repository. Participants were previously deployed active duty US military personnel (2002-2008) who had a recent archived serum sample available for analysis. Vitamin D status was estimated by measuring 25(OH) D levels in serum samples drawn within 24 months of the suicide. Each verified suicide case (n = 495) was matched to a control (n = 495) by rank, age and sex. We calculated odds ratio of suicide associated with categorical levels (octiles) of 25(OH) D, adjusted by season of serum collection. FINDINGS: More than 30% of all subjects had 25(OH)D values below 20 ng/mL. Although mean serum 25(OH)D concentrations did not differ between suicide cases and controls, risk estimates indicated that subjects in the lowest octile of season-adjusted 25(OH)D (<15.5 ng/mL) had the highest risk of suicide, with subjects in the subsequent higher octiles showing approximately the same level of decreased risk (combined odds ratio compared to lowest octile = 0.49; 95% C.I.: 0.315-0.768). CONCLUSIONS: Low vitamin D status is common in active duty service members. The lowest 25(OH)D levels are associated with an increased risk for suicide. Future studies could determine if additional sunlight exposure and vitamin D supplementation might reduce suicide by increasing 25(OH) D levels. PMCID: PMC3537724 PMID: 23308099 [PubMed - in process] 5. Prostaglandins Leukot Essent Fatty Acids. 2013 Jan;88(1):97-103. doi: 10.1016/j.plefa.2012.05.008. Epub 2012 Jul 4. n-3 Polyunsaturated fatty acids in animal models with neuroinflammation. Orr SK, Trépanier MO, Bazinet RP. Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada M5S 3E2. Neuroinflammation is present in the majority of acute and chronic neurological disorders. Excess or prolonged inflammation in the brain is thought to exacerbate neuronal damage and loss. Identifying modulators of neuroinflammation is an active area of study since it may lead to novel therapies. Omega-3 polyunsaturated fatty acids (n-3 PUFA) are anti-inflammatory in many non-neural tissues; their role in neuroinflammation is less studied. This review summarizes the relationship between n-3 PUFA and brain inflammation in animal models of brain injury and aging. Evidence by and large shows protective effects of n-3 PUFA in models of sickness behavior, stroke, aging, depression, Parkinson's disease, diabetes, and cytokine- and irradiation-induced cognitive impairments. However, rigorous studies that test the direct effects of n-3 PUFA in neuroinflammation in vivo are lacking. Future research in this area is necessary to determine if, and if so which, n-3 PUFA directly target brain inflammatory pathways. n-3 PUFA bioactive metabolites may provide novel therapeutic targets for neurological disorders with a neuroinflammatory component. Copyright © 2012 Elsevier Ltd. All rights reserved. PMID: 22770766 [PubMed - in process] 6. J Spec Oper Med. 2012 Fall;12(3):5-7. Omega-3 Fatty Acid Ingestion as a TBI Prophylactic. Barringer N, Conkright W. Given the hazardous nature of combat operations and training exercises (e.g. airborne operations) conducted by the United States military, servicemembers are at high risk for sustaining a traumatic brain injury (TBI). Since the beginning of the Iraq and Afghanistan wars, almost a quarter of a million servicemembers have sustained a TBI.1 A large number of TBIs are a result of the concussive forces generated by improvised explosive devices (IED). A smaller number are a result of penetrating head wounds. Others may be caused by activities resulting in powerful acceleration, deceleration, or rotational forces. Therapies for treating TBI thus far have been limited. Much of the research conducted to date has focused on post-injury pharmacological interventions.2 Additionally, better protective equipment could help in preventing TBIs; however, these issues are outside the scope of this paper. A relatively new area of research is investigating prophylactic measures taken to lessen the effects of TBI. One such measure involves nutritional interventions and their effects on TBI severity. Therefore, the purpose of this paper is to elucidate the potential benefits of omega-3 fatty acid intake as it relates to TBI severity. 2012. PMID: 23032313 [PubMed - in process] 7. Exp Transl Stroke Med. 2012 Sep 14;4(1):19. doi: 10.1186/2040-7378-4-19. Docosahexaenoic acid complexed to human albumin in experimental stroke: neuroprotective efficacy with a wide therapeutic window. Eady TN, Khoutorova L, Atkins KD, Bazan NG, Belayev L. Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA. lbelay@lsuhsc.edu. BACKGROUND: Docosahexaenoic acid (DHA) complexed to human serum albumin (Alb) is neuroprotective after experimental stroke. Here we tested using lower concentrations of albumin as part of the complex to achieve neuroprotection. We found that lower Alb concentrations extend the therapeutic window of protection beyond 5 h after stroke onset. METHODS: Sprague-Dawley rats were received 2 h middle cerebral artery occlusion (MCAo). The behavior was evaluated on day 1, 2, 3 and 7 after MCAo. In the dose-response study, animals were given either DHA (5mg/kg), Alb (0.63g/kg), DHA-Alb (5mg/kg + 0.32, 0.63 or 1.25 g/kg) or saline, i.v. 3 h after onset of stroke (n=6-8 per group). In the therapeutic window study, DHA-Alb (5mg/kg + 1.25g/kg) was administered i.v. at either 3, 4, 5, 6 or 7 h after onset of stroke (n=7-9 per group). Alb (1.25g/kg) was given at 3 h or 5 h and saline at 3h after onset of reperfusion. Seven days after MCAo, infarct volumes and number of GFAP, ED-1, NeuN, SMI-71 positive cells and vessels were counted. RESULTS: Moderate DHA-Alb doses (0.63 and 1.25 g/kg) improved neurological scores compared to albumin-treated rats on days 1, 2, 3 and 7. All DHA-Alb doses (0.32, 0.63 and 1.25 g/kg) markedly reduced cortical (by 65-70%), striatal (by 52-63%) and total infarct volumes (by 60-64%) compared to native Alb group. In the therapeutic window study DHA-Alb led to improved neurological score and significant reductions of infarct volumes (especially in the cortical or penumbral region), even when treatment was initiated as late as 7 hours after onset of MCAo. CONCLUSIONS: The DHA-Alb complex affords high-grade neurobehavioral neuroprotection in focal cerebral ischemia, equaling or exceeding that afforded by native Alb or DHA, at considerably moderate doses. It has a broad therapeutic window extending to 7 h after stroke onset. Taken together, these finding support the potential clinical feasibility of administering DHA-Alb therapy to patients with acute ischemic stroke. PMCID: PMC3540001 PMID: 22980673 [PubMed] 8. J Neurosurg Spine. 2012 Aug;17(2):134-40. doi: 10.3171/2012.5.SPINE1216. Epub 2012 Jun 26. Dietary therapy to promote neuroprotection in chronic spinal cord injury. Holly LT, Blaskiewicz D, Wu A, Feng C, Ying Z, Gomez-Pinilla F. Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA. lholly@mednet.ucla.edu OBJECT: The pathogenesis of cervical spondylotic myelopathy (CSM) is related to both primary mechanical and secondary biological injury. The authors of this study explored a novel, noninvasive method of promoting neuroprotection in myelopathy by using curcumin to minimize oxidative cellular injury and the capacity of omega-3 fatty acids to support membrane structure and improve neurotransmission. METHODS: An animal model of CSM was created using a nonresorbable expandable polymer placed in the thoracic epidural space, which induced delayed myelopathy. Animals that underwent placement of the expandable polymer were exposed to either a diet rich in docosahexaenoic acid and curcumin (DHA-Cur) or a standard Western diet (WD). Twenty-seven animals underwent serial gait testing, and spinal cord molecular assessments were performed after the 6-week study period. RESULTS: At the conclusion of the study period, gait analysis revealed significantly worse function in the WD group than in the DHA-Cur group. Levels of brain-derived neurotrophic factor (BDNF), syntaxin-3, and 4-hydroxynonenal (4-HNE) were measured in the thoracic region affected by compression and lumbar enlargement. Results showed that BDNF levels in the DHA-Cur group were not significantly different from those in the intact animals but were significantly greater than in the WD group. Significantly higher lumbar enlargement syntaxin-3 in the DHA-Cur animals combined with a reduction in lipid peroxidation (4-HNE) indicated a possible healing effect on the plasma membrane. CONCLUSIONS: Data in this study demonstrated that DHA-Cur can promote spinal cord neuroprotection and neutralize the clinical and biochemical effects of myelopathy. PMID: 22735048 [PubMed - indexed for MEDLINE] 9. Neurotoxicology. 2012 Aug;33(4):780-8. doi: 10.1016/j.neuro.2012.02.018. Epub 2012 Mar 8. n-3 polyunsaturated fatty acids inhibit lipopolysaccharide-induced microglial activation and dopaminergic injury in rats. Ji A, Diao H, Wang X, Yang R, Zhang J, Luo W, Cao R, Cao Z, Wang F, Cai T. Department of Nutrition & Food Hygiene and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China. Increasing evidence indicates that neuroinflammation plays an important role in neurotoxins-induced neurodegenerations. Microglia are a type of glial cells in the brain and play as the first and main form of active immune defense in the central nervous system. Accumulated data suggest that the activation of microglia plays a critical role in neurotoxicities induced by environmental toxicants. So the inhibition of microglia has been proven to be an effective strategy against neurotoxic effects. In the present study, we found that n-3 polyunsaturated fatty acids can inhibit both microglial activation and dopaminergic injury in the substantia nigra of Sprague-Dawley rats induced by lipopolysaccharide, one of the major constituents of the outer membrane of Gram-negative bacteria. Moreover, n-3 polyunsaturated fatty acids inhibited lipopolysaccharide-induced activation of nuclear factor-κB, an important transcription factor involved in microglial activation. Taken together, our results provided the first in vivo evidence that n-3 polyunsaturated fatty acids can inhibit the damage of dopaminergic neurons induced by lipopolysaccharide through their inhibitory effects on nuclear factor-κB-dependent microglial activation. Copyright © 2012 Elsevier Inc. All rights reserved. PMID: 22406923 [PubMed - indexed for MEDLINE] 10. Exp Neurol. 2012 Jul;236(1):122-30. doi: 10.1016/j.expneurol.2012.04.007. Epub 2012 Apr 19. Novel aspirin-triggered neuroprotectin D1 attenuates cerebral ischemic injury after experimental stroke. Bazan NG, Eady TN, Khoutorova L, Atkins KD, Hong S, Lu Y, Zhang C, Jun B, Obenaus A, Fredman G, Zhu M, Winkler JW, Petasis NA, Serhan CN, Belayev L. Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA. nbazan@lsuhsc.edu Acute ischemic stroke triggers complex neurovascular, neuroinflammatory and synaptic alterations. Aspirin and docosahexaenoic acid (DHA), an omega-3 essential fatty acid family member, have beneficial effects on cerebrovascular diseases. DHA is the precursor of neuroprotectin D1 (NPD1), which downregulates apoptosis and, in turn, promotes cell survival. Here we have tested the effect of aspirin plus DHA administration and discovered the synthesis of aspirin-triggered NPD1 (AT-NPD1) in the brain. Then we performed the total chemical synthesis of this molecule and tested in the setting of 2h middle cerebral artery occlusion (MCAo) in Sprague-Dawley rats. Neurological status was evaluated at 24h, 48 h, 72 h, and 7 days. At 3h post-stroke onset, an intravenous administration of 333 μg/kg of AT-NPD1 sodium salt (AT-NPD1-SS) or methyl-ester (AT-NPD1-ME) or vehicle (saline) as treatment was given. On day 7, ex vivo magnetic resonance imaging (MRI) of the brains was conducted on 11.7 T MRI. T2WI, 3D volumes, and apparent diffusion coefficient (ADC) maps were generated. In addition, infarct volumes and number of GFAP (reactive astrocytes), ED-1 (activated microglia/macrophages) and SMI-71-positive vessels were counted in the cortex and striatum at the level of the central lesion. All animals showed similar values for rectal and cranial temperatures, arterial blood gases, and plasma glucose during and after MCAo. Treatment with both AT-NPD1-SS and AT-NPD1-ME significantly improved neurological scores compared to saline treatment at 24h, 48 h, 72 h and 7 days. Total lesion volumes computed from T2WI images were significantly reduced by both AT-NPD1-SS and AT-NPD1-ME treatment in the cortex (by 44% and 81%), striatum (by 61% and 77%) and total infarct (by 48% and 78%, respectively). Brain edema, computed from T2WI in the cortex (penumbra) and striatum (core), was elevated in the saline group. In contrast, both AT-NPD1 decreased water content in the striatum on day 7. 3D volumes, computed from T2WI, were dramatically reduced with both AT-NPD1 and the lesion was mostly localized in the subcortical areas. Treatment with both AT-NPD1-SS and AT-NPD1-ME significantly reduced cortical (by 76% and 96%), subcortical (by 61% and 70%) and total (69% and 84%, respectively) infarct volumes as defined by histopathology. In conclusion, a novel biosynthetic pathway that leads to the formation of AT-NPD1 mediator in the brain was discovered. In addition, administration of synthetic AT-NPD1, in either its sodium salt or as the methyl ester, was able to attenuate cerebral ischemic injury which leads to a novel approach for pharmaceutical intervention and clinical translation. Copyright © 2012 Elsevier Inc. All rights reserved. PMCID: PMC3409566 [Available on 2013/7/1] PMID: 22542947 [PubMed - indexed for MEDLINE] 11. Int J Dev Neurosci. 2012 Jun;30(4):315-23. doi: 10.1016/j.ijdevneu.2012.01.007. Epub 2012 Jan 28. Maternal prenatal omega-3 fatty acid supplementation attenuates hyperoxia-induced apoptosis in the developing rat brain. Tuzun F, Kumral A, Ozbal S, Dilek M, Tugyan K, Duman N, Ozkan H. Department of Neonatology, School of Medicine, Dokuz Eylul University, Turkey. Supraphysiologic amounts of oxygen negatively influences brain maturation and development. The aim of the present study was to evaluate whether maternal ω-3 long-chain polyunsaturated fatty acid (ω-3 FA) supplementation during pregnancy protects the developing brain against hyperoxic injury. Thirty-six rat pups from six different dams were divided into six groups according to the diet modifications and hyperoxia exposure. The groups were: a control group (standard diet+room air), a hyperoxia group (standard diet+80% O₂ exposure), a hyperoxia+high-dose ω-3 FA-supplemented group, a hyperoxia+low-dose ω-3 FA-supplemented group, a room air+low-dose ω-3 FA-supplemented+group, and a room air+high dose ω-3 FA-supplemented group. The ω-3 FA's were supplemented as a mixture of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) from the second day of pregnancy until birth. Rat pups in the hyperoxic groups were exposed to 80% oxygen from birth until postnatal day 5 (P5). At P5, all animals were sacrificed. Neuronal cell death and apoptosis were evaluated by cell count, TUNEL, and active Caspase-3 immunohistochemistry. Histopathological examination showed that maternally ω-3 FA deficient diet and postnatal hyperoxia exposure were associated with significantly lower neuronal counts and significantly higher apoptotic cell death in the selected brain regions. Ω-3 FA treatment significantly diminished apoptosis, in the selected brain regions, in a dose dependent manner. Our results suggest that the maternal ω-3 FA supply may protect the developing brain against hyperoxic injury. Copyright © 2012 ISDN. Published by Elsevier Ltd. All rights reserved. PMID: 22342579 [PubMed - indexed for MEDLINE] 12. J Matern Fetal Neonatal Med. 2012 Jun;25(6):849-54. doi: 10.3109/14767058.2011.587917. Epub 2011 Sep 6. Maternal omega-3 fatty acid supplementation protects against lipopolysaccharide-induced white matter injury in the neonatal rat brain. Tuzun F, Kumral A, Dilek M, Ozbal S, Ergur B, Yesilirmak DC, Duman N, Yilmaz O, Ozkan H. Department of Pediatrics, Division of Neonatology, Dokuz Eylul University, Izmir, Turkey. OBJECTIVES: Periventricular leukomalacia (PVL) is the predominant form of brain injury in premature infants, and no specific treatment currently exists for this condition. We have evaluated whether maternal omega-3 fatty acid (ω3 FA) treatment reduces endotoxin-induced PVL in the developing rat brain. METHODS: Wistar rats with dated pregnancies were fed a standard diet or a diet enriched in ω3 FA (70% docosahexaenoic acid + 30% eicosapentaenoic acid mixture) during gestation. Intraperitoneal injection of lipopolysaccharide (LPS) was administered consecutively on the 18th and 19th embryonic days to establish the endotoxin-induced PVL rat model. The animals were divided into four groups: (i) control, (ii) PVL, (iii) PVL+low-dose ω3 FA and (iv) PVL+high-dose ω3 FA. At day P7, apoptosis and hypomyelination in periventricular white matter were evaluated by immunohistochemical assessments. RESULTS: High-dose maternal ω3 FA treatment reduced brain weight loss. Maternal ω3 FA treatment given either in low or high doses greatly decreased caspase-3 immunoreactivity and increased myelin basic protein immunoreactivity, indicating a decrease in apoptosis and hypomyelination. CONCLUSION: Considering that no specific treatment is available for PVL, maternal ω3 FA supplementation may provide a nutritional strategy to limit periventricular white matter damage caused by infections during pregnancy. PMID: 21892882 [PubMed - indexed for MEDLINE] 13. J Neurochem. 2012 Jun;121(5):738-50. doi: 10.1111/j.1471-4159.2012.07726.x. Epub 2012 Apr 12. Docosahexaenoic acid, but not eicosapentaenoic acid, reduces the early inflammatory response following compression spinal cord injury in the rat. Hall JC, Priestley JV, Perry VH, Michael-Titus AT. Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA. jodie.hall@osumc.edu Docosahexaenoic acid (DHA, 22 : 6) and eicosapentaenoic acid (EPA, 20 : 5) are omega-3 polyunsaturated fatty acids (n-3 PUFAs) with distinct anti-inflammatory properties. Both have neuroprotective effects acutely following spinal cord injury (SCI). We examined the effect of intravenous DHA and EPA on early inflammatory events after SCI. Saline, DHA or EPA (both 250 nmol/kg) were administered 30 min after T12 compression SCI, to female Sprague-Dawley rats. DHA significantly reduced the number of neutrophils to some areas of the injured epicentre at 4 h and 24 h. DHA also reduced C-reactive protein plasma levels, whereas EPA did not significantly reduce neutrophils or C-reactive protein. Laminectomy and SCI elicited a sustained inflammatory response in the liver, which was not reversed by the PUFAs. The chemokine KC/GRO/CINC and the cytokine IL-6 provide gradients for chemotaxis of neutrophils to the epicentre. At 4 h after injury, there was a significant increase in IL-6, KC/GRO/CINC, IL-1β and tumour necrosis factor-α in the epicentre, with a return to baseline at 24 h. Neither DHA nor EPA returned their levels to control values. These results indicate that the acute neuroprotective effects of n-3 PUFAs in rat compression SCI may be only partly attributed to reduction of some of the early inflammatory events occurring after injury. © 2012 The Authors. Journal of Neurochemistry © 2012 International Society for Neurochemistry. PMID: 22404382 [PubMed - indexed for MEDLINE] 14. Stroke. 2012 Mar;43(3):691-6. doi: 10.1161/STROKEAHA.111.636035. Epub 2012 Jan 26. Free Fatty acids and delayed cerebral ischemia after subarachnoid hemorrhage. Badjatia N, Seres D, Carpenter A, Schmidt JM, Lee K, Mayer SA, Claassen J, Connolly ES, Elkind MS. Neurological Institute of New York, 177 Fort Washington Avenue, New York, NY 10032, USA. nb2217@columbia.edu BACKGROUND AND PURPOSE: The purpose of this study was to understand factors related to increases in serum free fatty acid (FFA) levels and association with delayed cerebral ischemia (DCI) after subarachnoid hemorrhage. METHODS: We performed serial measurement of systemic oxygen consumption by indirect calorimetry and FFA levels by liquid chromatography/mass spectrometry in the first 14 days after ictus in 50 consecutive patients with subarachnoid hemorrhage. Multivariable generalized estimating equation models identified associations with FFA levels in the first 14 days after SAH and Cox proportional hazards model used to identified associations with time to DCI. RESULTS: There were 187 measurements in 50 patients with subarachnoid hemorrhage (mean age, 56±14 years old; 66% women) with a median Hunt-Hess score of 3. Adjusting for Hunt-Hess grade and daily caloric intake, n-6 and n-3 FFA levels were both associated with oxygen consumption and the modified Fisher score. Fourteen (28%) patients developed DCI on median postbleed Day 7. The modified Fisher score (P=0.01), mean n-6:n-3 FFA ratio (P=0.02), and mean oxygen consumption level (P=0.04) were higher in patients who developed DCI. In a Cox proportional hazards model, the mean n-6:n-3 FFA ratio (P<0.001), younger age (P=0.05), and modified Fisher scale (P=0.004) were associated with time to DCI. CONCLUSIONS: Injury severity and oxygen consumption hypermetabolism are associated with higher n-FFA levels and an increased n-6:n-3 FFA ratio is associated with DCI. This may indicate a role for interventions that modulate both oxygen consumption and FFA levels to reduce the occurrence of DCI. PMID: 22282893 [PubMed - indexed for MEDLINE] 15. Transl Stroke Res. 2012 Mar;3(1):154-163. Epub 2011 Oct 27. Superior Neuroprotective Efficacy of LAU-0901, a Novel Platelet-Activating Factor Antagonist, in Experimental Stroke. Belayev L, Eady TN, Khoutorova L, Atkins KD, Obenaus A, Cordoba M, Vaquero JJ, Alvarez-Builla J, Bazan NG. Platelet-activating factor (PAF) accumulates during cerebral ischemia, and inhibition of this process plays a critical role in neuronal survival. Recently, we demonstrated that LAU-0901, a novel PAF receptor antagonist, is neuroprotective in experimental stroke. We used magnetic resonance imaging in conjunction with behavior and immunohistopathology to expand our understanding of this novel therapeutic approach. Sprague-Dawley rats received 2 h middle cerebral artery occlusion (MCAo) and were treated with LAU-0901 (60 mg/kg) or vehicle 2 h from MCAo onset. Behavioral function, T2-weighted imaging (T2WI), and apparent diffusion coefficients were performed on days 1, 3, and 7 after MCAo. Infarct volume and number of GFAP, ED-1, and NeuN-positive cells were conducted on day 7. Behavioral deficit was significantly improved by LAU-0901 treatment compared to vehicle on days 1, 3, and 7. Total lesion volumes computed from T2WI were significantly reduced by LAU-0901 on days 1, 3, and 7 (by 83%, 90%, and 96%, respectively), which was consistent with decreased edema formation. Histopathology revealed that LAU-0901 treatment resulted in significant reduction of cortical and subcortical infarct volumes, attenuated microglial infiltration, and promoted astrocytic and neuronal survival. These findings suggest LAU-0901 is a promising neuroprotectant and provide the basis for future therapeutics in patients suffering ischemic stroke. PMCID: PMC3284672 PMID: 22408693 [PubMed] 16. PLoS One. 2012;7(10):e46151. doi: 10.1371/journal.pone.0046151. Epub 2012 Oct 30. Docosahexaenoic acid signaling modulates cell survival in experimental ischemic stroke penumbra and initiates long-term repair in young and aged rats. Eady TN, Belayev L, Khoutorova L, Atkins KD, Zhang C, Bazan NG. Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America. BACKGROUND: Docosahexaenoic acid, a major omega-3 essential fatty acid family member, improves behavioral deficit and reduces infarct volume and edema after experimental focal cerebral ischemia. We hypothesize that DHA elicits neuroprotection by inducing AKT/p70S6K phosphorylation, which in turn leads to cell survival and protects against ischemic stroke in young and aged rats. METHODS AND RESULTS: Rats underwent 2 h of middle cerebral artery occlusion (MCAo). DHA, neuroprotectin D1 (NPD1) or vehicle (saline) was administered 3 h after onset of stroke. Neurological function was evaluated on days 1, 2, 3, and 7. DHA treatment improved functional recovery and reduced cortical, subcortical and total infarct volumes 7 days after stroke. DHA also reduced microglia infiltration and increased the number of astrocytes and neurons when compared to vehicle on days 1 and 7. Increases in p473 AKT and p308 AKT phosphorylation/activation were observed in animals treated with DHA 4 h after MCAo. Activation of other members of the AKT signaling pathway were also observed in DHA treated animals including increases in pS6 at 4 h and pGSK at 24 h. DHA or NPD1 remarkably reduced total and cortical infarct in aged rats. Moreover, we show that in young and aged rats DHA treatment after MCAo potentiates NPD1 biosynthesis. The phosphorylation of p308 AKT or pGSK was not different between groups in aged rats. However, pS6 expression was increased with DHA or NPD1 treatment when compared to vehicle. CONCLUSIONS: We suggest that DHA induces cell survival, modulates the neuroinflammatory response and triggers long term restoration of synaptic circuits. Both DHA and NPD1 elicited remarkable protection in aged animals. Accordingly, activation of DHA signaling might provide benefits in the management of ischemic stroke both acutely as well as long term to limit ensuing disabilities. PMCID: PMC3484151 PMID: 23118851 [PubMed - in process] 17. J Clin Psychiatry. 2011 Dec;72(12):1585-90. doi: 10.4088/JCP.11m06879. Epub 2011 Aug 23. Suicide deaths of active-duty US military and omega-3 fatty-acid status: a case-control comparison. Lewis MD, Hibbeln JR, Johnson JE, Lin YH, Hyun DY, Loewke JD. Department of Preventive Medicine and Biometrics, Uniformed Services University of the Health Sciences Bethesda, Maryland, USA. Comment in J Clin Psychiatry. 2011 Dec;72(12):1574-6. BACKGROUND: The recent escalation of US military suicide deaths to record numbers has been a sentinel for impaired force efficacy and has accelerated the search for reversible risk factors. OBJECTIVE: To determine whether deficiencies of neuroactive, highly unsaturated omega-3 essential fatty acids (n-3 HUFAs), in particular docosahexaenoic acid (DHA), are associated with increased risk of suicide death among a large random sample of active-duty US military. METHOD: In this retrospective case-control study, serum fatty acids were quantified as a percentage of total fatty acids among US military suicide deaths (n = 800) and controls (n = 800) matched for age, date of collection of sera, sex, rank, and year of incident. Participants were active-duty US military personnel (2002-2008). For cases, age at death ranged from 17-59 years (mean = 27.3 years, SD = 7.3 years). Outcome measures included death by suicide, postdeployment health assessment questionnaire (Department of Defense Form 2796), and ICD-9 mental health diagnosis data. RESULTS: Risk of suicide death was 14% higher per SD of lower DHA percentage (OR = 1.14; 95% CI, 1.02-1.27; P < .03) in adjusted logistic regressions. Among men, risk of suicide death was 62% greater with low serum DHA status (adjusted OR = 1.62; 95% CI, 1.12-2.34; P < .01, comparing DHA below 1.75% [n = 1,389] to DHA of 1.75% and above [n = 141]). Risk of suicide death was 52% greater in those who reported having seen wounded, dead, or killed coalition personnel (OR = 1.52; 95% CI, 1.11-2.09; P < .01). CONCLUSION: This US military population had a very low and narrow range of n-3 HUFA status. Although these data suggest that low serum DHA may be a risk factor for suicide, well-designed intervention trials are needed to evaluate causality. © Copyright 2011 Physicians Postgraduate Press, Inc. PMCID: PMC3259251 PMID: 21903029 [PubMed - indexed for MEDLINE] 18. J Trauma. 2011 Nov;71(5):1211-8. doi: 10.1097/TA.0b013e3182092c62. Lipid mediators in cerebral spinal fluid of traumatic brain injured patients. Farias SE, Heidenreich KA, Wohlauer MV, Murphy RC, Moore EE. Department of Pharmacology, University of Colorado at Denver, Anschutz Medical Campus, Aurora, CO, USA. BACKGROUND: Enzymatic and nonenzymatic oxidation of polyunsaturated fatty acids leads to the formation of biologically active products known as lipid mediators. In the brain, lipid mediators play an important role in supporting homeostasis and normal function. Thus, levels of these metabolites in normal and pathologic conditions in the brain are particularly relevant in understanding the transition to disease. METHODS: In this study, liquid chromatography tandem mass spectrometry was used to analyze lipid mediators in cerebrospinal fluid (CSF) of controls and traumatic brain injured (TBI) patients. RESULTS: Our results showed that the levels of arachidonic acid (AA), docosahexaenoic acid (DHA), 5- and 12- eicosatetraenoic acid (HETE) were significantly increased in the CSF of TBI patients. The magnitude of increase was 10-fold for AA, DHA, and 5-HETE and 17-fold for 12-HETE. Prostaglandins and leukotrienes were not detected in CSF of either control or brain injured patients. Furthermore, this study found that isoprostanes and thromboxanes are present in CSF of brain injured patients. CONCLUSIONS: This study clearly shows that certain lipid mediators accumulate in the CSF of TBI patient. This study also suggests the potential use of DHA, AA, 5- and 12-HETE as biochemical markers of brain injury and to monitor the impact of interventions. PMID: 21427623 [PubMed - indexed for MEDLINE] 19. J Neurotrauma. 2011 Oct;28(10):2113-22. doi: 10.1089/neu.2011.1872. Epub 2011 Oct 4. The salutary effects of DHA dietary supplementation on cognition, neuroplasticity, and membrane homeostasis after brain trauma. Wu A, Ying Z, Gomez-Pinilla F. Department of Integrative Biology and Physiology, University of California at Los Angeles (UCLA), Los Angeles, California 90095, USA. The pathology of traumatic brain injury (TBI) is characterized by the decreased capacity of neurons to metabolize energy and sustain synaptic function, likely resulting in cognitive and emotional disorders. Based on the broad nature of the pathology, we have assessed the potential of the omega-3 fatty acid docosahexaenoic acid (DHA) to counteract the effects of concussive injury on important aspects of neuronal function and cognition. Fluid percussion injury (FPI) or sham injury was performed, and rats were then maintained on a diet high in DHA (1.2% DHA) for 12 days. We found that DHA supplementation, which elevates brain DHA content, normalized levels of brain-derived neurotrophic factor (BDNF), synapsin I (Syn-1), cAMP-responsive element-binding protein (CREB), and calcium/calmodulin-dependent kinase II (CaMKII), and improved learning ability in FPI rats. It is known that BDNF facilitates synaptic transmission and learning ability by modulating Syn-I, CREB, and CaMKII signaling. The DHA diet also counteracted the FPI-reduced manganese superoxide dismutase (SOD) and Sir2 (a NAD+-dependent deacetylase). Given the involvement of SOD and Sir2 in promoting metabolic homeostasis, DHA may help the injured brain by providing resistance to oxidative stress. Furthermore, DHA normalized levels of calcium-independent phospholipase A2 (iPLA2) and syntaxin-3, which may help preserve membrane homeostasis and function after FPI. The overall results emphasize the potential of dietary DHA to counteract broad and fundamental aspects of TBI pathology that may translate into preserved cognitive capacity. PMCID: PMC3191367 PMID: 21851229 [PubMed - indexed for MEDLINE] 20. Mil Med. 2011 Oct;176(10):1120-7. Neuroprotection for the warrior: dietary supplementation with omega-3 fatty acids. Lewis MD, Bailes J. Defense and Veterans Brain Injury Center, 11300 Rockville Pike Suite 1100, Rockville, MD 20812, USA. Nutrition has traditionally involved in supplying energy and hydration. An emerging concept developed by the authors is the concept of using omega-3 fatty acids (n-3 FAs) to increase the resilience of the brain. The n-3 FAs have numerous proven benefits including support of cardiovascular and psychiatric health. Docosahexaenoic acid in particular, is found in high concentrations in the brain. N-3 FAs provide benefits by exerting a protective mechanism at the cellular and neuronal levels including the modulation of inflammatory cascade following traumatic brain injury. Promising research and evolving clinical experience now indicate that n-3 FA is useful and effective for recovery following traumatic brain injury. More exciting is that new laboratory research shows the beneficial effects extend to when n-3 FA is given before injury. Given the safety profile, availability, and affordability of n-3 FA, Generally Recognized As Safe amounts of eicosapentaenoic acid and docosahexaenoic acid (up to 3,000 mg daily) should be considered for the athlete and soldier, not only for its general health benefits, but particularly also for those at risk or high exposure to brain impacts. A comprehensive, coordinated research program to evaluate the multiple uses of n-3 FA should be a high priority for the Department of Defense. PMID: 22128646 [PubMed - indexed for MEDLINE] 21. Mol Neurobiol. 2011 Oct;44(2):216-22. doi: 10.1007/s12035-011-8200-6. Epub 2011 Sep 15. Endogenous signaling by omega-3 docosahexaenoic acid-derived mediators sustains homeostatic synaptic and circuitry integrity. Bazan NG, Musto AE, Knott EJ. Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA, nbazan@lsuhsc.edu. The harmony and function of the complex brain circuits and synapses are sustained mainly by excitatory and inhibitory neurotransmission, neurotrophins, gene regulation, and factors, many of which are incompletely understood. A common feature of brain circuit components, such as dendrites, synaptic membranes, and other membranes of the nervous system, is that they are richly endowed in docosahexaenoic acid (DHA), the main member of the omega-3 essential fatty acid family. DHA is avidly retained and concentrated in the nervous system and known to play a role in neuroprotection, memory, and vision. Only recently has it become apparent why the surprisingly rapid increases in free (unesterified) DHA pool size take place at the onset of seizures or brain injury. This phenomenon began to be clarified by the discovery of neuroprotectin D1 (NPD1), the first-uncovered bioactive docosanoid formed from free DHA through 15-lipoxygenase-1 (15-LOX-1). NPD1 synthesis includes, as agonists, oxidative stress and neurotrophins. The evolving concept is that DHA-derived docosanoids set in motion endogenous signaling to sustain homeostatic synaptic and circuit integrity. NPD1 is anti-inflammatory, displays inflammatory resolving activities, and induces cell survival, which is in contrast to the pro-inflammatory actions of the many of omega-6 fatty acid family members. We highlight here studies relevant to the ability of DHA to sustain neuronal function and protect synapses and circuits in the context of DHA signalolipidomics. DHA signalolipidomics comprises the integration of the cellular/tissue mechanism of DHA uptake, its distribution among cellular compartments, the organization and function of membrane domains containing DHA phospholipids, and the precise cellular and molecular events revealed by the uncovering of signaling pathways regulated by docosanoids endowed with prohomeostatic and cell survival bioactivity. Therefore, this approach offers emerging targets for prevention, pharmaceutical intervention, and clinical translation involving DHA-mediated signaling. PMCID: PMC3180614 PMID: 21918832 [PubMed - indexed for MEDLINE] 22. Stroke. 2011 Oct;42(10):2903-9. doi: 10.1161/STROKEAHA.111.620856. Epub 2011 Aug 18. Accumulation of dietary docosahexaenoic acid in the brain attenuates acute immune response and development of postischemic neuronal damage. Lalancette-Hébert M, Julien C, Cordeau P, Bohacek I, Weng YC, Calon F, Kriz J. Department of Psychiatry and Neuroscience, Faculty of Medicine, University Laval, 2705 boul. Laurier, Quebec City, QC, G1V 4G2 Canada. BACKGROUND AND PURPOSE: Consumption of fish has been shown to reduce risk of coronary heart disease and, possibly, of ischemic stroke. Because docosahexaenoic acid (DHA) is the most likely neuroactive component within fish oil, we hypothesized that exposing mice to a DHA-enriched diet may reduce inflammation and protect neurons against ischemic injury. METHODS: To visualize the effects of DHA on neuroinflammation after stroke, TLR2-fluc-GFP transgenic mice were exposed to either a control diet, a diet depleted in n-3 polyunsaturated fatty acid, or a diet enriched in DHA during 3 months. Real-time biophotonic/bioluminescence imaging of the TLR2 response was performed before and after middle cerebral artery occlusion, whereas cytokines concentrations and stroke area analyses were performed at 3 and 7 days after middle cerebral artery occlusion, respectively. RESULTS: We show that a 3-month DHA treatment prevented microglial activation after ischemic injury, reduced the ischemic lesion size, and increased levels of the antiapoptotic molecule Bcl-2 in the brain. Additional analysis revealed a significant decrease in the levels of COX2 and IL-1β, but not in other proinflammatory cytokines. Importantly, long-term DHA supplementation significantly changed the n-3:n-6 polyunsaturated fatty acid ratio in the brain. CONCLUSIONS: Collectively, these data indicate that diet-induced accumulation of DHA in the brain protects against postischemic inflammation and injury. Because DHA is widely available at low cost and has an excellent safety profile, our data suggest that increased DHA intake may provide protection against acute immune response/brain damage in ischemic stroke. PMID: 21852616 [PubMed - indexed for MEDLINE] 23. JPEN J Parenter Enteral Nutr. 2011 Sep;35(5):556-9. doi: 10.1177/0148607111416122. Nutrition and traumatic brain injury: a perspective from the Institute of Medicine report. Bistrian BR, Askew W, Erdman JW Jr, Oria MP. Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA. bbistria@bidmc.harvard.edu PMID: 21881010 [PubMed - indexed for MEDLINE] 24. Chem Biol. 2011 Aug 26;18(8):976-87. doi: 10.1016/j.chembiol.2011.06.008. Novel proresolving aspirin-triggered DHA pathway. Serhan CN, Fredman G, Yang R, Karamnov S, Belayev LS, Bazan NG, Zhu M, Winkler JW, Petasis NA. Center for Experimental Therapeutics and Reperfusion Injury, Harvard Institutes of Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, USA. cnserhan@zeus.bwh.harvard.edu Comment in Chem Biol. 2011 Oct 28;18(10):1208-9. Endogenous mechanisms in the resolution of acute inflammation are of interest because excessive inflammation underlies many pathologic abnormalities. We report an aspirin-triggered DHA metabolome that biosynthesizes a potent product in inflammatory exudates and human leukocytes, namely aspirin-triggered Neuroprotectin D1/Protectin D1 [AT-(NPD1/PD1)]. The complete stereochemistry of AT-(NPD1/PD1) proved to be 10R,17R-dihydroxydocosa-4Z,7Z,11E,13E,15Z,19Z-hexaenoic acid. The chirality of hydroxyl groups and geometry of the conjugated triene system essential for bioactivity were established by matching biological materials with stereochemically pure isomers prepared by organic synthesis. AT-(NPD1/PD1) reduced neutrophil (PMN) recruitment in murine peritonitis in a dose-dependent fashion whereby neither a Δ(15)-trans-isomer nor DHA was effective. With human cells, AT-(NPD1/PD1) decreased transendothelial PMN migration as well as enhanced efferocytosis of apoptotic human PMN by macrophages. These results indicate that AT-(NPD1/PD1) is a potent anti-inflammatory proresolving molecule. Copyright © 2011 Elsevier Ltd. All rights reserved. PMCID: PMC3164791 PMID: 21867913 [PubMed - indexed for MEDLINE] 25. Neurosci Lett. 2011 Jun 8;496(3):168-71. doi: 10.1016/j.neulet.2011.04.009. Epub 2011 Apr 14. Oral fish oil restores striatal dopamine release after traumatic brain injury. Shin SS, Dixon CE. Brain Trauma Research Center, Department of Neurosurgery, University of Pittsburgh, 3434 Fifth Ave, Suite 201, Pittsburgh, PA 15260, USA. Omega-3 fatty acid administration can affect the release of neurotransmitters and reduce inflammation and oxidative stress, but its use in traumatic brain injury (TBI) has not been described extensively. We investigated the effect of 7 day oral fish oil treatment in the recovery of potassium evoked dopamine release after TBI. Sham rats and TBI rats were given either olive oil or fish oil by oral gavage and were subject to cerebral microdialysis. Olive oil treated TBI rats showed significant dopamine release deficit compared to sham rats, and this deficit was restored with oral fish oil treatment. There was no effect of fish oil treatment on extracellular levels of dopamine metabolites such as 3,4-dihydroxyphenylacetic acid and homovanillic acid. These results suggest the therapeutic potential of omega-3 fatty acids in restoring dopamine neurotransmission deficits after TBI. Published by Elsevier Ireland Ltd. PMID: 21514362 [PubMed - indexed for MEDLINE] 26. Prev Med. 2011 Jun;52 Suppl 1:S75-80. doi: 10.1016/j.ypmed.2011.01.023. Epub 2011 Jan 31. The combined effects of exercise and foods in preventing neurological and cognitive disorders. Gomez-Pinilla F. Dept. of Integrative Biology and Physiology, and Dept. of Neurosurgery, University of California Los Angeles, Los Angeles, CA 90095, USA. fgomezpi@ucla.edu OBJECTIVE: Exercise and select diets have important influences on health and plasticity of the nervous system, and the molecular mechanisms involved with these actions are starting to be elucidated. New evidence indicates that exercise, in combination with dietary factors, exerts its effects by affecting molecular events related to the management of energy metabolism and synaptic plasticity. METHODS: Published studies in animals and humans describing the effects of exercise and diets in brain plasticity and cognitive abilities are discussed. RESULTS: New evidence indicates that exercise and select diets exert their effects by affecting molecular events related to the management of energy metabolism and synaptic plasticity. An important instigator in the molecular machinery stimulated by exercise is brain-derived neurotrophic factor (BDNF), which acts at the interface of metabolism and plasticity. CONCLUSIONS: Recent studies show that selected dietary factors share similar mechanisms with exercise, and in some cases they can complement the action of exercise. Therefore, exercise and dietary management appear as a non-invasive and effective strategy to counteract neurological and cognitive disorders. Copyright © 2011 Elsevier Inc. All rights reserved. PMCID: PMC3258093 PMID: 21281667 [PubMed - indexed for MEDLINE] 27. J Stroke Cerebrovasc Dis. 2011 May-Jun;20(3):188-95. doi: 10.1016/j.jstrokecerebrovasdis.2009.11.016. Epub 2010 Jul 10. Eicosapentaenoic acid prevents memory impairment after ischemia by inhibiting inflammatory response and oxidative damage. Okabe N, Nakamura T, Toyoshima T, Miyamoto O, Lu F, Itano T. Department of Neurobiology, Kagawa University Faculty of Medicine, 1750-1 Ikenobe, Miki, Kagawa, Japan. Previous studies have demonstrated that the generation of reactive oxygen species and an excessive inflammatory reaction are involved in the progression of neural damage following brain ischemia. In this study, we focused on the anti-inflammatory and antioxidant properties of eicosapentaenoic acid (EPA). Gerbils were treated intraperitoneally with 500 mg/kg of EPA ethyl for 4 weeks until the day of forebrain ischemia, which was induced by occluding the bilateral common carotid artery for 5 minutes. In the first part of the 2-part experiment, the effect of EPA treatment was evaluated using hematoxylin and eosin staining and deoxynucleotidyl transferase-mediated dUTP nick-end labeling as a marker of cell death (n=3 per group). The inflammatory reaction was evaluated using anti-Iba1 immunohistochemistry, a marker of microglial activation (n=3 per group), and detection of 8-hydroxyl-2'-deoxyguanosine, a marker of oxidative DNA damage (n=4 per group). In the second part of the experiment, the effect of EPA treatment on memory function was examined using an 8-arm radial maze (n=6 per group). EPA treatment significantly inhibited DNA oxidative damage (P < .05) and accumulation of Iba1-positive cells in the CA1 area at 12 and 72 hours after the induction of ischemia, and also decreased apoptotic neurons and neuronal death (P < .001) at 72 hours after ischemia. EPA treatment also significantly improved memory function (P < .05). These findings suggest that EPA inhibits the inflammatory reaction and oxidative damage occurring after ischemic brain injury, and also may contribute to the prevention of neural damage and memory impairment following such injury. Copyright © 2011 National Stroke Association. Published by Elsevier Inc. All rights reserved. PMID: 20621517 [PubMed - indexed for MEDLINE] 28. Curr Opin Clin Nutr Metab Care. 2011 Mar;14(2):158-67. doi: 10.1097/MCO.0b013e328342cba5. Docosahexaenoic acid: brain accretion and roles in neuroprotection after brain hypoxia and ischemia. Mayurasakorn K, Williams JJ, Ten VS, Deckelbaum RJ. Institute of Human Nutrition, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York 10032, USA. PURPOSE OF REVIEW: With important effects on neuronal lipid composition, neurochemical signaling and cerebrovascular pathobiology, docosahexaenoic acid (DHA), a n-3 polyunsaturated fatty acid, may emerge as a neuroprotective agent against cerebrovascular disease. This paper examines pathways for DHA accretion in brain and evidence for possible roles of DHA in prophylactic and therapeutic approaches for cerebrovascular disease. RECENT FINDINGS: DHA is a major n-3 fatty acid in the mammalian central nervous system and enhances synaptic activities in neuronal cells. DHA can be obtained through diet or to a limited extent via conversion from its precursor, α-linolenic acid (α-LNA). DHA attenuates brain necrosis after hypoxic ischemic injury, principally by modulating membrane biophysical properties and maintaining integrity in functions between presynaptic and postsynaptic areas, resulting in better stabilizing intracellular ion balance in hypoxic-ischemic insult. Additionally, DHA alleviates brain apoptosis, by inducing antiapoptotic activities such as decreasing responses to reactive oxygen species, upregulating antiapoptotic protein expression, downregulating apoptotic protein expression, and maintaining mitochondrial integrity and function. SUMMARY: DHA in brain relates to a number of efficient delivery and accretion pathways. In animal models DHA renders neuroprotection after hypoxic-ischemic injury by regulating multiple molecular pathways and gene expression. PMID: 21178607 [PubMed - indexed for MEDLINE] 29. Transl Stroke Res. 2011 Mar;2(1):33-41. Epub 2010 Nov 4. Docosahexaenoic Acid Therapy of Experimental Ischemic Stroke. Belayev L, Khoutorova L, Atkins KD, Eady TN, Hong S, Lu Y, Obenaus A, Bazan NG. We examined the neuroprotective efficacy of docosahexaenoic acid (DHA), an omega-3 essential fatty acid family member, in acute ischemic stroke; studied the therapeutic window; and investigated whether DHA administration after an ischemic stroke is able to salvage the penumbra. In each series described below, SD rats underwent 2 h of middle cerebral artery occlusion (MCAo). In series 1, DHA or saline was administered i.v. at 3, 4, 5, or 6 h after stroke. In series 2, MRI was conducted on days 1, 3 and 7. In series 3, DHA or saline was administered at 3 h, and lipidomic analysis was conducted on day 3. Treatment with DHA significantly improved behavior and reduced total infarct volume by a mean of 40% when administered at 3 h, by 66% at 4 h, and by 59% at 5 h. Total lesion volumes computed from T2-weighted images were reduced in the DHA group at all time points. Lipidomic analysis showed that DHA treatment potentiates neuroprotectin D1 (NPD1) synthesis in the penumbra 3 days after MCAo. DHA administration provides neurobehavioral recovery, reduces brain infarction and edema, and activates NPD1 synthesis in the penumbra when administered up to 5 h after focal cerebral ischemia in rats. PMCID: PMC3037476 PMID: 21423332 [PubMed] 30. Neurosurgery. 2011 Feb;68(2):474-81; discussion 481. doi: 10.1227/NEU.0b013e3181ff692b. Dietary supplementation with the omega-3 fatty acid docosahexaenoic acid in traumatic brain injury. Mills JD, Hadley K, Bailes JE. Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, West Virginia, USA. BACKGROUND: Although various strategies for prevention of brain disease have been implemented, no substance has been found to be advantageous for prophylaxis against brain injury. OBJECTIVE: While previous work in our laboratory and others have shown positive effects using the omega-3 fatty acid docosahexaenoic acid (DHA) in post-injury treatment following traumatic and ischemic insults, we wished to test its effects when given prior to injury. We have attempted to measure anatomical, cellular, and behavioral outcomes with a prophylactic administration of DHA. METHODS: Five groups of 16 adult male Sprague-Dawley rats were subjected to an impact acceleration traumatic brain after having received a prior administration of DHA in doses of 3, 12, and 40 mg/kg for 30 days prior. Serum fatty acid levels were determined from isolated plasma phospholipids at baseline and at the end of 30 days supplementation. Following sacrifice 1 week after injury, brainstem white matter tracts underwent fluorescent immunohistochemical processing for labeling of beta amyloid precursor protein (APP), an anatomical marker of brain injury, as well as measurements of CD68 and caspase-3 levels, and water maze testing was used for behavioral assessment. RESULTS: Dietary supplementation with DHA resulted in increased serum DHA levels proportionate with the escalating dosage. Immunohistochemical analysis revealed significantly (P < .05) decreased numbers of APP levels in all groups of animals receiving pre-injury supplementation with DHA of 4, 12, and 40 mg/kg at 13955, 4186, and 2827 axons per mm³, respectively, vs 37442 in unsupplemented animals, as measured by stereological methodology. Using a selective measuring technique, only the highest dosage group, 40 mg/kg showed significantly (P < .05) decreased numbers of APP positive axons, at 1.15 axons per high power field vs 6.78 in unsupplemented animals. CD-68, caspase-3, and water maze testing all were significantly (P < .05) improved in the high dose group. CONCLUSION: Dietary supplementation with DHA increases serum levels and, if given prior to traumatic brain injury, reduces the injury response, as measured by axonal injury counts, markers for cellular injury and apoptosis, and memory assessment by water maze testing. This uniform response was seen for the highest dosage group, 40 mg/kg given over 30 days prior to injury, but when measured by stereological counting methodology there was a positive response to anatomical injury across low to high doses of DHA. The potential for DHA to provide prophylactic benefit to the brain against traumatic injury appears promising and requires further investigation. PMID: 21135750 [PubMed - indexed for MEDLINE] 31. Acta Cir Bras. 2011;26 Suppl 1:32-7. Preconditioning with oil mixes of high ratio Omega-9: Omega-6 and a low ratio Omega-6:Omega-3 in rats subjected to brain ischemia/reperfusion. Pinheiro PM, Campelo AP, Guimarães SB, Patrocínio RM, Valença Junior JT, Vasconcelos PR. Department of Surgery, Federal University of Ceara, Fortaleza-CE, Brazil. PURPOSE: This study aimed to assess the effects of preconditioning with mixtures of oils containing high/low ratio of ω-6/ω-3 and ω-9/ω-6, respectively, in an experimental model of cerebral ischemia-reperfusion (I/R). METHODS: Forty-two Wistar rats were randomly distributed into two groups: control (n=24) and test (n=18). Control group was subdivided in 4 subgroups (n=6): G1: Sham-Water; G2: I/R-Water; G3: Sham-Isolipidic and G4: I/R-Isolipid. The animals received water or a isolipid mixture containing ω-3 oils (8:1 ratio) and ω-9/ω-6 (0.4:1 ratio) by gavage for seven days. Test group included 3 subgroups (n=6) G5: I/R-Mix1, G: 6 I/R-Mix2 and G7: I/R-Mix3. Test group animals received oily mixtures of ω-3 (1.4:1 ratio) and ω-6 (3.4:1 ratio), differing only in source of ω-3: G5 (alpha-linolenic acid); G6 (alpha-linolenic, docosahexaenoic and eicosapentaenoic acids), and G7 (alpha-linolenic and docosahexaenoic acids). On day 7 I/R rats underwent cerebral ischemia with bilateral occlusion of common carotid arteries for 1 hour followed by reperfusion for 3 hours. G1 and G3 animals underwent sham operation. Concluded the experiment, animals were decapitated and their brains sliced for red neurons (RN) count in CA3 area of the hippocampus. Variables were compared using ANOVA-Tukey test. RESULTS: The use of different mix preparations promoted a decrease in red cell count in all three groups (G5/G6/G7), compared with G2/G4, confirming the protective effect of different oil blends, regardless of ω-3 source. CONCLUSION: Pre-conditioning with mixtures of oils containing high ratio ω-6/ω-3 and low ω-9/ω-6 relationship protects brain neurons against I/R injury in an experimental model. PMID: 21971654 [PubMed - indexed for MEDLINE] 32. Clin Neurosurg. 2011;58:51-6. Concussion management at the NFL, college, high school, and youth sports levels. Maroon JC, Bost J. University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA. PMID: 21916127 [PubMed - indexed for MEDLINE] 33. J Neurosurg. 2011 Jan;114(1):77-84. doi: 10.3171/2010.5.JNS08914. Epub 2010 Jul 16. Omega-3 fatty acid supplementation and reduction of traumatic axonal injury in a rodent head injury model. Mills JD, Bailes JE, Sedney CL, Hutchins H, Sears B. Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, West Virginia, USA. jmills@hsc.wvu.edu OBJECT: Traumatic brain injury remains the most common cause of death in persons under 45 years of age in the Western world. Recent evidence from animal studies suggests that supplementation with omega-3 fatty acid (O3FA) (particularly eicosapentaenoic acid [EPA] and docosahexaenoic acid [DHA]) improves functional outcomes following focal neural injury. The purpose of this study is to determine the benefits of O3FA supplementation following diffuse axonal injury in rats. METHODS: Forty adult male Sprague-Dawley rats were used. Three groups of 10 rats were subjected to an impact acceleration injury and the remaining group underwent a sham-injury procedure (surgery, but no impact injury). Two of the groups subjected to the injury were supplemented with 10 or 40 mg/kg/day of O3FA; the third injured group served as an unsupplemented control group. The sham-injured rats likewise received no O3FA supplementation. Serum fatty acid levels were determined from the isolated plasma phospholipids prior to the injury and at the end of the 30 days of supplementation. After the animals had been killed, immunohistochemical analysis of brainstem white matter tracts was performed to assess the presence of β-amyloid precursor protein (APP), a marker of axonal injury. Immunohistochemical analyses of axonal injury mechanisms-including analysis for caspase-3, a marker of apoptosis; RMO-14, a marker of neurofilament compaction; and cytochrome c, a marker of mitochondrial injury-were performed. RESULTS: Dietary supplementation with a fish oil concentrate rich in EPA and DHA for 30 days resulted in significant increases in O3FA serum levels: 11.6% ± 4.9% over initial levels in the 10 mg/kg/day group and 30.7% ± 3.6% in the 40 mg/kg/day group. Immunohistochemical analysis revealed significantly (p < 0.05) decreased numbers of APP-positive axons in animals receiving O3FA supplementation: 7.7 ± 14.4 axons per mm(2) in the 10 mg/kg/day group and 6.2 ± 11.4 axons per mm(2) in the 40 mg/kg/day group, versus 182.2 ± 44.6 axons per mm(2) in unsupplemented animals. Sham-injured animals had 4.1 ± 1.3 APP-positive axons per mm(2). Similarly, immunohistochemical analysis of caspase-3 expression demonstrated significant (p < 0.05) reduction in animals receiving O3FA supplementation, 18.5 ± 28.3 axons per mm(2) in the 10 mg/kg/day group and 13.8 ± 18.9 axons per mm(2) in the 40 mg/kg/day group, versus 129.3 ± 49.1 axons per mm(2) in unsupplemented animals. CONCLUSIONS: Dietary supplementation with a fish oil concentrate rich in the O3FAs EPA and DHA increases serum levels of these same fatty acids in a dose-response effect. Omega-3 fatty acid supplementation significantly reduces the number of APP-positive axons at 30 days postinjury to levels similar to those in uninjured animals. Omega-3 fatty acids are safe, affordable, and readily available worldwide to potentially reduce the burden of traumatic brain injury. PMID: 20635852 [PubMed - indexed for MEDLINE] 34. Nutr Health. 2011;20(3-4):165-9. Collaborative effects of diet and exercise on cognitive enhancement. Gomez-Pinilla F. Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA. fgomezpi@ucla.edu Certain dietary factors, such as omega-3 fatty acids and curcumin, are reviewed in their context of stimulating molecular systems that serve synaptic function, while diets rich in saturated fats do the opposite. In turn, exercise, using similar mechanisms as healthy diets, displays healing effects on the brain such as counteracting the mental decline associated with age and facilitating functional recovery resulting from brain injury and disease. Diet and exercise are two noninvasive approaches that used together may enhance neural repair. Omega 3 fatty acids and curcumin elevate levels of molecules important for synaptic plasticity such as brain-derived neurotrophic factor (BDNF), thus benefiting normal brain function and recovery events following brain insults. PMCID: PMC3258096 PMID: 22141190 [PubMed - indexed for MEDLINE] 35. Neuropathology. 2010 Dec;30(6):597-605. doi: 10.1111/j.1440-1789.2010.01114.x. Maternal docosahexaenoic acid-enriched diet prevents neonatal brain injury. Suganuma H, Arai Y, Kitamura Y, Hayashi M, Okumura A, Shimizu T. Department of Pediatrics, Juntendo University School of Medicine, Tokyo, Japan. hsuganu@med.juntendo.ac.jp Hypoxic-ischemic encephalopathy due to neonatal asphyxia is one of the most important causes of delayed neurological development. Prolonged neuronal apoptosis plays an important role in the processes contributing to neuronal degeneration. Docosahexaenoic acid (DHA), a major component of brain membrane phospholipids, prevents neuronal cell apoptosis and plays an important role as an anti-oxidant agent. We investigated the neuroprotective and anti-oxidant effects of maternal DHA supplementation during pregnancy in a model of neonatal hypoxic-ischemic encephalopathy. Pregnant rats were randomly assigned to two experimental groups: a control group or a DHA-enriched diet group. Hypoxic-ischemic encephalopathy was produced by left common carotid artery occlusion and exposure to 8% oxygen for 1.5 h. TUNEL assay, immunohistochemistry for caspase-3 and 8-hydroxy-deoxyguanosine (8-OHdG), and Western blot for caspase-3 were performed at postnatal days 8, 10 and 14. Fatty acid composition of brain was estimated on postnatal day 7. Maternal diet clearly influenced brain fatty acid composition in pups. Numbers of apoptotic neuronal cells and 8-OHdG immunoreactivity were significantly decreased in the DHA-enriched group. Our findings indicate that maternal DHA-enriched diet during pregnancy provides neuroprotection by inhibiting oxidative stress and apoptotic neuronal death. Dietary supplementation of DHA during pregnancy may thus be beneficial in preventing neonatal brain injury. © 2010 Japanese Society of Neuropathology. PMID: 20408962 [PubMed - indexed for MEDLINE] 36. Stroke. 2010 Oct;41(10):2341-7. doi: 10.1161/STROKEAHA.110.586081. Epub 2010 Aug 12. Omega-3 polyunsaturated fatty acid supplementation confers long-term neuroprotection against neonatal hypoxic-ischemic brain injury through anti-inflammatory actions. Zhang W, Hu X, Yang W, Gao Y, Chen J. Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA. chenj2@upmc.edu BACKGROUND AND PURPOSE: Current available therapies for neonatal hypoxia/ischemia (H/I) brain injury are rather limited. Here, we investigated the effect of omega-3 polyunsaturated fatty acids on brain damage and long-term neurological function after H/I in neonates. METHODS: Female rats were treated with or without an omega-3 polyunsaturated fatty acids-enriched diet from the second day of pregnancy until 14 days after parturition. Seven-day-old neonates were subjected to H/I and euthanized 5 weeks later for evaluation of tissue loss. Neurological impairment was assessed progressively for 5 weeks after H/I by grid walking, foot fault, and Morris water maze. Activation of microglia and production of inflammatory mediators were examined up to 7 days after H/I. RESULTS: Omega-3 polyunsaturated fatty acid supplementation significantly reduced brain damage and improved long-term neurological outcomes up to 5 weeks after neonatal H/I injury. Omega-3 polyunsaturated fatty acids exerted an anti-inflammatory effect in microglia both in an in vivo model of H/I and in in vitro microglial cultures subjected to inflammatory stimuli by inhibiting NF-κB activation and subsequent release of inflammatory mediators. CONCLUSIONS: Our results suggest that omega-3 polyunsaturated fatty acids confer potent neuroprotection against neonatal H/I brain injury through, at least partially, suppressing a microglial-mediated inflammatory response. PMCID: PMC3021248 PMID: 20705927 [PubMed - indexed for MEDLINE] 37. J Neurotrauma. 2010 Sep;27(9):1617-24. doi: 10.1089/neu.2009.1239. Docosahexaenoic acid reduces traumatic axonal injury in a rodent head injury model. Bailes JE, Mills JD. Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, West Virginia 26506, USA. jbailes@hsc.wvu.edu Traumatic brain injury (TBI) remains the most common cause of death in persons under age 45 in the Western world. Recent evidence from animal studies suggests that supplementation with omega-3 fatty acids (O3FA) improves functional outcomes following focal neural injury. The purpose of this study is to determine the benefits of DHA supplementation following diffuse axonal injury in rats. Four groups of 10 adult male Sprague-Dawley rats were subjected to an impact acceleration injury and then received 30 days of supplementation with either 10 mg/kg/d or 40 mg/kg/d of docosahexaenoic acid (DHA). Serum fatty acid levels were determined from the isolated plasma phospholipids prior to injury and at the end of the 30 days of DHA supplementation. Following sacrifice, brainstem white matter tracts underwent fluorescent immunohistochemical processing for labeling of β-amyloid precursor protein (APP), a marker of axonal injury. Dietary supplementation with either 10 mg/kg/d or 40 mg/kg/d of DHA for 30 days results in significantly (p < 0.05) increased DHA serum levels of 123% and 175% over baseline, respectively. Immunohistochemical analysis reveals significantly (p < 0.05) decreased numbers of APP-positive axons in animals receiving dietary supplementation with DHA, 26.1 (SD 5.3) for 10 mg/kg/d, and 19.6 (SD 4.7) for 40 mg/kg/d axons per mm(2), versus 147.7 (SD 7.1) axons in unsupplemented animals. Sham-injured animals had 6.4 (SD 13.9) APP positive axons per mm(2). Dietary supplementation with DHA increases serum levels in a dose-dependent manner. DHA supplementation significantly reduces the number of APP-positive axons at 30 days post-injury, to levels similar to seen those in uninjured animals. DHA is safe, affordable, and readily available worldwide to potentially reduce the burden of TBI. PMID: 20597639 [PubMed - indexed for MEDLINE] 38. Mol Neurobiol. 2010 Jun;41(2-3):367-74. doi: 10.1007/s12035-010-8139-z. Epub 2010 May 14. Omega-3 essential fatty acids modulate initiation and progression of neurodegenerative disease. Palacios-Pelaez R, Lukiw WJ, Bazan NG. Diater Laboratorios, 28918, Leganes, Madrid, Spain. r.palacios@diater.com The significance of the selective enrichment in omega-3 essential fatty acids in photoreceptors and synaptic membranes of the nervous system has remained, until recently, incompletely understood. While studying mechanisms of cell survival in neural degeneration, we discovered a docosanoid synthesized from unesterified docosahexaenoic acid (DHA) by a 15-lipoxygenase (15-LOX), which we called neuroprotectin D1 (NPD1; 10R,17S-dihydroxy-docosa-4Z,7Z,11E,13E,15E,19Z hexaenoic acid). This lipid mediator is a docosanoid because it is derived from the 22 carbon (22C) precursor DHA, unlike eicosanoids, which are derived from the 20 carbon (20C) arachidonic acid (AA) family member of essential fatty acids. We discovered that NPD1 is promptly made in response to oxidative stress, as a response to brain ischemia-reperfusion, and in the presence of neurotrophins. NPD1 is neuroprotective in experimental brain damage, in oxidative-stressed retinal pigment epithelial (RPE) cells, and in human brain cells exposed to amyloid-beta (Abeta) peptides. We thus envision NPD1 as a protective sentinel, one of the very first defenses activated when cell homeostasis is threatened by imbalances in normal neural function. We provide here, in three sections, recent experimental examples that highlight the specificity and potency of NPD1 spanning beneficial bioactivity during initiation and early progression of neurodegeneration: (1) during retinal signal phototransduction, (2) during brain ischemia-reperfusion, and (3) in Alzheimer's disease (AD) and stressed human brain cell models of AD. From this experimental evidence, we conclude that DHA-derived NPD1 regulation targets upstream events of brain cell apoptosis, as well as neuro-inflammatory signaling, promoting and maintaining cellular homeostasis, and restoring neural and retinal cell integrity. PMID: 20467837 [PubMed - indexed for MEDLINE] 39. Am J Obstet Gynecol. 2010 May;202(5):469.e1-6. doi: 10.1016/j.ajog.2010.01.076. Epub 2010 Mar 31. Docosahexaenoic acid confers neuroprotection in a rat model of perinatal hypoxia-ischemia potentiated by Escherichia coli lipopolysaccharide-induced systemic inflammation. Berman DR, Liu YQ, Barks J, Mozurkewich E. Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI. OBJECTIVE: Lipopolysaccharide pretreatment potentiates hypoxic ischemic injury. We hypothesized that docosahexaenoic acid pretreatment would improve function and reduce brain volume loss in this rat model of perinatal brain injury and inflammation. STUDY DESIGN: Seven-day-old rats were divided into 3 groups: intraperitoneal docosahexaenoic acid 1 mg/kg and lipopolysaccharide 0.1 mg/kg, 25% albumin and lipopolysaccharide, and normal saline. Injections were given 2.5 hours before carotid ligation, followed by 90 minutes 8% O2. Rats underwent sensorimotor function testing and brain volume loss assessment on postnatal day 14. RESULTS: Docosahexaenoic acid pretreatment improved vibrissae forepaw placing scores compared with albumin/lipopolysaccharide (mean+/-standard deviation weighted score/20: 17.72+/-0.92 docosahexaenoic acid/lipopolysaccharide vs 13.83+/-0.82 albumin/lipopolysaccharide; P<.007). Albumin/lipopolysaccharide rats scores were worse than those of the normal saline/normal saline rats (13.83+/-0.82 vs 17.21+/-0.71; P=.076). No significant differences in brain volume loss were observed among groups. CONCLUSION: Lipopolysaccharide inflammatory stimulation in conjunction with hypoxic ischemic resulted in poorer function than hypoxic ischemic alone. Docosahexaenoic acid pretreatment had significantly improved function in neonatal rats exposed to lipopolysaccharide and hypoxic ischemic. Copyright (c) 2010 Mosby, Inc. All rights reserved. PMCID: PMC3535291 PMID: 20356570 [PubMed - indexed for MEDLINE] 40. Prostaglandins Other Lipid Mediat. 2010 Apr;91(3-4):85-9. doi: 10.1016/j.prostaglandins.2009.09.005. Epub 2009 Oct 3. Docosahexaenoic acid neurolipidomics. Niemoller TD, Bazan NG. Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, School of Medicine, New Orleans, LA 70112, USA. Mediator lipidomics is a field of study concerned with the characterization, structural elucidation and bioactivity of lipid derivatives actively generated by enzymatic activity. It is well known that omega-3 fatty acids are beneficial for brain function. Docosahexaenoic acid [DHA; 4 22:6(n-3)] is the most abundant essential omega-3 fatty acid present in the brain and it has multiple mechanisms of exerting protective effects after cellular injury. Certain lipid species produced from DHA early during the reperfusion stage of brain ischemia-reperfusion injury are generated in order to help the cell cope as the injury progresses. We explore these newly discovered lipid mediators in order to understand their role in the cell. We have identified one of these potentially protective lipid mediators as a novel stereospecific DHA-derived fatty acid, called neuroprotectin D1 (NPD1; 10R,17S-dihydroxy-docosa-4Z,7Z,11E,15E,19Z hexaenoic acid). DHA also has important roles in pro-survival signaling cascades after ischemia-reperfusion in injury. It has been shown to accelerate AKT translocation and activation and has binding affinity with an important PPAR-gamma family of ligand-activated nuclear receptors that have been implicated in various aspects of lipid metabolism and have been shown to have anti-inflammatory actions. Here we present an overview of these mechanisms and discuss the potential of using DHA signaling in the development of treatments for the large population of patients suffering from the devastating consequences of stroke. Copyright 2009. Published by Elsevier Inc. PMCID: PMC2905848 PMID: 19804838 [PubMed - indexed for MEDLINE] 41. Acta Neurol Taiwan. 2009 Dec;18(4):231-41. Essential fatty acids and human brain. Chang CY, Ke DS, Chen JY. Department of Neurology, Chi-Mei Medical Center, Tainan Taiwan. chiayu.chang7@msa.hinet.net The human brain is nearly 60 percent fat. We've learned in recent years that fatty acids are among the most crucial molecules that determine your brain's integrity and ability to perform. Essential fatty acids (EFAs) are required for maintenance of optimal health but they can not synthesized by the body and must be obtained from dietary sources. Clinical observation studies has related imbalance dietary intake of fatty acids to impaired brain performance and diseases. Most of the brain growth is completed by 5-6 years of age. The EFAs, particularly the omega-3 fatty acids, are important for brain development during both the fetal and postnatal period. Dietary decosahexaenoic acid (DHA) is needed for the optimum functional maturation of the retina and visual cortex, with visual acuity and mental development seemingly improved by extra DHA. Beyond their important role in building the brain structure, EFAs, as messengers, are involved in the synthesis and functions of brain neurotransmitters, and in the molecules of the immune system. Neuronal membranes contain phospholipid pools that are the reservoirs for the synthesis of specific lipid messengers on neuronal stimulation or injury. These messengers in turn participate in signaling cascades that can either promote neuronal injury or neuroprotection. The goal of this review is to give a new understanding of how EFAs determine our brain's integrity and performance, and to recall the neuropsychiatric disorders that may be influenced by them. As we further unlock the mystery of how fatty acids affect the brain and better understand the brain's critical dependence on specific EFAs, correct intake of the appropriate diet or supplements becomes one of the tasks we undertake in pursuit of optimal wellness. PMID: 20329590 [PubMed - indexed for MEDLINE] 42. J Nutr Biochem. 2009 Sep;20(9):715-25. doi: 10.1016/j.jnutbio.2008.06.014. Epub 2008 Sep 20. Protective effect of docosahexaenoic acid against brain injury in ischemic rats. Pan HC, Kao TK, Ou YC, Yang DY, Yen YJ, Wang CC, Chuang YH, Liao SL, Raung SL, Wu CW, Chiang AN, Chen CJ. Chung Hwa University of Medical Technology, Tainan, Taiwan. Evidence suggests that inactivation of cell-damaging mechanisms and/or activation of cell-survival mechanisms may provide effective preventive or therapeutic interventions to reduce cerebral ischemia/reperfusion (I/R) injuries. Docosahexaenoic acid (DHA) is an essential polyunsaturated fatty acid in the central nervous system that has been shown to possess neuroprotective effects. We examined whether different preadministrative protocols of DHA have effects on brain injury after focal cerebral I/R and investigated the potential neuroactive mechanisms involved. Sprague-Dawley rats were intraperitoneally pretreated with DHA once 1 h or 3 days being subjected to focal cerebral I/R or daily for 6 weeks before being subjected to focal cerebral I/R. Reduction of brain infarction was found in all three DHA-pretreated groups. The beneficial effect of DHA on the treatment groups was accompanied by decreases in blood-brain barrier disruption, brain edema, malondialdehyde (MDA) production, inflammatory cell infiltration, interleukin-6 (IL-6) expression and caspase-3 activity. Elevation of antioxidative capacity, as evidenced by decreased MDA level and increased superoxide dismutase activity and glutathione level, was detected only in the chronic daily-administration group. The two single-administration groups showed increased phosphorylation of extracellular-signal-regulated kinase (ERK). Elevation of Bcl-2 expression was detected in the chronic daily-administration and 3-day-administration groups. In vitro study demonstrated that DHA attenuated IL-6 production from stimulated glial cells involving nuclear factor kappaB inactivation. Therefore, the data suggest that the neuroprotective mechanisms of DHA pretreatment are, in part, mediated by attenuating damaging mechanisms through reduction of cytotoxic factor production and by strengthening survival mechanisms through ERK-mediated and/or Bcl-2-mediated prosurvival cascade. PMID: 18805685 [PubMed - indexed for MEDLINE] 43. Stroke. 2009 Sep;40(9):3121-6. doi: 10.1161/STROKEAHA.109.555979. Epub 2009 Jun 18. Robust docosahexaenoic acid-mediated neuroprotection in a rat model of transient, focal cerebral ischemia. Belayev L, Khoutorova L, Atkins KD, Bazan NG. Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA. lbelay@lsuhsc.edu BACKGROUND AND PURPOSE: Docosahexaenoic acid (DHA; 22:6n-3), an omega-3 essential fatty acid family member, is the precursor of neuroprotectin D1, which downregulates apoptosis and, in turn, promotes cell survival. This study was conducted to assess whether DHA would show neuroprotective efficacy when systemically administered in different doses after middle cerebral artery occlusion (MCAo) in rats. METHODS: Sprague-Dawley rats were anesthetized with isoflurane and subjected to 2 hour of MCAo. Animals were treated with either DHA (low doses=3.5 or 7 mg/kg; medium doses=16 or 35 mg/kg; and high dose=70 mg/kg) or an equivalent volume of saline intravenously 3 hours after MCAo onset. Neurologic status was evaluated during occlusion (60 minutes) and on days 1, 2, 3, and 7 after MCAo. Seven days after MCAo, brains were perfusion-fixed, and infarct areas and volumes were determined. RESULTS: Only the low and medium doses of DHA significantly improved the neurologic score compared with vehicle-treated rats at 24 hours, 48 hours, 72 hours, and 7 days. DHA markedly reduced total corrected infarct volume in all treated groups compared with vehicle-treated rats (3.5 mg/kg, 26+/-9 mm(3); 7 mg/kg, 46+/-12 mm(3); 16 mg/kg, 37+/-5 mm(3); and 35 mg/kg, 34+/-15 mm(3) vs vehicle, 94+/-12 mm(3)). Cortical and striatal infarct volumes were also significantly reduced by treatment with DHA. No neuroprotective effects were observed with 70 mg/kg DHA. CONCLUSIONS: We conclude that DHA experimental therapy at low and medium doses improves neurologic and histologic outcomes after focal cerebral ischemia and might provide benefits in patients after ischemic stroke. PMCID: PMC2745047 PMID: 19542051 [PubMed - indexed for MEDLINE] 44. Prostaglandins Leukot Essent Fatty Acids. 2009 Aug-Sep;81(2-3):205-11. doi: 10.1016/j.plefa.2009.05.024. Epub 2009 Jun 11. Cellular and molecular events mediated by docosahexaenoic acid-derived neuroprotectin D1 signaling in photoreceptor cell survival and brain protection. Bazan NG. Neuroscience Center of Excellence and Department of Ophthalmology, School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite D, New Orleans, LA 70112, USA. nbazan@lsuhsc.edu Deficiency in docosahexaenoic acid (DHA) is associated with impaired visual and neurological postnatal development, cognitive decline, macular degeneration, and other neurodegenerative diseases. DHA is an omega-3 polyunsaturated fatty acyl chain concentrated in phospholipids of brain and retina, with photoreceptor cells displaying the highest content of DHA of all cell membranes. The identification and characterization of neuroprotectin D1 (NPD1, 10R, 17S-dihydroxy-docosa-4Z,7Z,11E,13E,15Z,19Z-hexaenoic acid) contributes in understanding the biological significance of DHA. In oxidative stress-challenged human retinal pigment epithelial (RPE) cells, human brain cells, or rat brains undergoing ischemia-reperfusion, NPD1 synthesis is enhanced as a response for sustaining homeostasis. Thus, neurotrophins, Abeta peptide 42 (Abeta42), calcium ionophore A23187, interleukin (IL)-1beta, or DHA supply enhances NPD1 synthesis. NPD1, in turn, up-regulates the antiapoptotic proteins of the Bcl-2 family and decreases the expression of proapoptotic Bcl-2 family members. Moreover, NPD1 inhibits IL-1beta-stimulated expression of cyclooxygenase-2 (COX-2). Because both RPE and photoreceptors are damaged and then die in retinal degenerations, elucidating how NPD1 signaling contributes to retinal cell survival may lead to a new understanding of disease mechanisms. In human neural cells, DHA attenuates amyloid-beta (Abeta) secretion, resulting in concomitant formation of NPD1. NPD1 was found to be reduced in the Alzheimer's disease (AD) cornu ammonis region 1 (CA1) hippocampal region, but not in other areas of the brain. The expression of key enzymes for NPD1 biosynthesis, cytosolic phospholipase A(2) (cPLA(2)), and 15-lipoxygenase (15-LOX) was found altered in the AD hippocampal CA1 region. NPD1 repressed Abeta42-triggered activation of pro-inflammatory genes and upregulated the antiapoptotic genes encoding Bcl-2, Bcl-xl, and Bfl-1(A1) in human brain cells in culture. Overall, these results support the concept that NPD1 promotes brain and retina cell survival via the induction of antiapoptotic and neuroprotective gene-expression programs that suppress Abeta42-induced neurotoxicity and other forms of cell injury, which in turn fosters homeostasis during development in aging, as well as during the initiation and progression of neurodegenerative diseases. PMCID: PMC2756692 PMID: 19520558 [PubMed - indexed for MEDLINE] 45. Altern Med Rev. 2009 Mar;14(1):14-35. Integrated brain restoration after ischemic stroke--medical management, risk factors, nutrients, and other interventions for managing inflammation and enhancing brain plasticity. Kidd PM. Brain injury from ischemic stroke can be devastating, but full brain restoration is feasible. Time until treatment is critical; rapid rate of injury progression, logistical and personnel constraints on neurological and cardiovascular assessment, limitations of recombinant tissue plasminogen activator (rtPA) for thrombolysis, anticoagulation and antiplatelet interventions, and neuroprotection all affect outcome. Promising acute neuroprotectant measures include albumin, magnesium, and hypothermia. Long-term hyperbaric oxygen therapy (HBOT) is safe and holds great promise. Eicosanoid and cytokine down-regulation by omega-3 nutrients docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) may help quench stroke inflammation. C-reactive protein (CRP), an inflammatory biomarker and stroke-recurrence predictor, responds favorably to krill oil (a phospholipid-DHA/EPA-astaxanthin complex). High homocysteine (Hcy) is a proven predictor of stroke recurrence and responds to folic acid and vitamin B12. Vitamin E may lower recurrence for individuals experiencing high oxidative stress. Citicoline shows promise for acute neuroprotection. Glycerophosphocholine (GPC) is neuroprotective and supports neuroplasticity via nerve growth factor (NGF) receptors. Stem cells have shown promise for neuronal restoration in randomized trials. Endogenous brain stem cells can migrate to an ischemic injury zone; exogenous stem cells once transplanted can migrate (home) to the stroke lesion and provide trophic support for cortical neuroplasticity. The hematopoietic growth factors erythropoietin (EPO) and granulocyte-colony stimulating factor (G-CSF) have shown promise in preliminary trials, with manageable adverse effects. Physical and mental exercises, including constraint-induced movement therapy (CIMT) and interactive learning aids, further support brain restoration following ischemic stroke. Brain plasticity underpins the function-driven brain restoration that can occur following stroke. PMID: 19364191 [PubMed - indexed for MEDLINE] 46. Brain Res. 2009 Feb 9;1253:184-90. doi: 10.1016/j.brainres.2008.11.074. Epub 2008 Dec 3. LAU-0901, a novel platelet-activating factor receptor antagonist, confers enduring neuroprotection in experimental focal cerebral ischemia in the rat. Belayev L, Khoutorova L, Atkins K, Cherqui A, Alvarez-Builla J, Bazan NG. Neuroscience Center of Excellence, Louisiana State University Health Science Center, New Orleans, LA 70112, USA. lbelay@lsuhsc.edu LAU-0901, a novel platelet-activating factor (PAF) receptor antagonist, is highly neuroprotective in a rodent model of cerebral ischemia. This study was conducted to establish whether the neuroprotection induced by LAU-0901 persists with chronic survival. Male Sprague-Dawley rats were anesthetized with isoflurane and subjected to 2 h of temporary middle cerebral artery occlusion (MCAo) induced by means of a poly-L-lisine-coated intraluminal nylon suture. Animals were treated with either LAU-0901 (60 mg/kg) or vehicle (45% cyclodextran) administered i.p. at 2 h from onset of MCAo. They received neurobehavioral examinations during MCAo (60 min) and then at 1, 2, 3, 7, 14, 21 and 28 days followed by histopathology at 30 days. LAU-0901 significantly improved the behavior compared to the vehicle group, beginning on day 1 (by 29%, p=0.00007) and persisting throughout a 30-day survival period (42%, p=0.0001). Compared with vehicle treatment, LAU-0901 treatment significantly increased volume of non-infarcted brain tissue loss relative to the unlesioned hemisphere (16.3 +/- 4.6% vs. 46.0 +/- 10.3%, respectively). These results establish that LAU-0901 confers enduring ischemic neuroprotection. PMCID: PMC2637461 PMID: 19070607 [PubMed - indexed for MEDLINE] 47. World Rev Nutr Diet. 2009;99:46-54. doi: 10.1159/000192994. Epub 2009 Jan 9. Omega-3 fatty acid docosahexaenoic acid is the precursor of neuroprotectin D1 in the nervous system. Niemoller TD, Stark DT, Bazan NG. Neuroscience Center of Excellence and Department of Ophthalmology, Louisiana State University Health Sciences Center, School of Medicine, New Orleans, La 70112, USA. PMID: 19136838 [PubMed - indexed for MEDLINE] 48. Exp Neurol. 2008 Dec;214(2):253-8. doi: 10.1016/j.expneurol.2008.08.009. Epub 2008 Aug 28. LAU-0901, a novel platelet-activating factor antagonist, is highly neuroprotective in cerebral ischemia. Belayev L, Khoutorova L, Atkins K, Gordon WC, Alvarez-Builla J, Bazan NG. Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA. lbelay@lsuhsc.edu Platelet-activating factor (PAF) is a bioactive phospholipid that accumulates during ischemia-reperfusion and is involved in the activation of platelets, neutrophils, and pro-inflammatory signaling. PAF has been suggested to enhance brain ischemia-reperfusion damage. LAU-0901, a novel PAF receptor antagonist, was examined in models of focal cerebral ischemia in rats and mice. Sprague-Dawley rats were anesthetized and received 2-hour middle cerebral artery occlusion (MCAo) by intraluminal suture. LAU-0901 (30, 60, 90 mg/kg; n=9-11) or vehicle (n=11) was administered i.p. at 2 h after onset of MCAo. The neurological status was evaluated at 60 min, and on days 1, 2, 3 and 7 after MCAo. In the dose-response study in mice, C57BL/6 mice were anesthetized and received 1 h MCAo by intraluminal suture. LAU-0901 (15, 30, 60 mg/kg; n=7-9) or vehicle (n=8) was given i.p. at 1 h after onset of MCAo. Local cerebral blood flow (LCBF) was measured at 1, 2, 4, and 6 h after MCAo in mice. LAU-0901 treated rats showed improved neurological score throughout the 7-day survival period. LAU-0901 treatment (30, 60 and 90 mg/kg) reduced total corrected infarct volume compared to vehicle rats by 76, 88 and 90%, respectively. Mice treated with LAU-0901 (30 and 60 mg/kg) reduced total infarction by 29% and 66%, respectively. LCBF was improved by treatment with LAU-0901 (30 mg/kg) by 77% of baseline at 6 h. In conclusion, we demonstrate for the first time that LAU-0901 improves behavioral scores, LCBF and reduces infarct volume after focal cerebral ischemia in rats and mice. Thus, this PAF receptor antagonist exhibits potent and sustained neuroprotection that may be of value for the design of stroke therapies. PMCID: PMC2647717 PMID: 18793637 [PubMed - indexed for MEDLINE] 49. J Neurotrauma. 2008 Dec;25(12):1499. Omega-3 and BDNF regulation: eicosapentaenoic acid may play a key role in limitation of CNS injury. Logan AC. Comment on J Neurotrauma. 2004 Oct;21(10):1457-67. PMID: 19378461 [PubMed - indexed for MEDLINE] 50. Epilepsy Behav. 2008 Jul;13(1):36-42. doi: 10.1016/j.yebeh.2008.01.001. Epub 2008 Mar 4. Neuroprotective activity of omega-3 fatty acids against epilepsy-induced hippocampal damage: Quantification with immunohistochemical for calcium-binding proteins. Ferrari D, Cysneiros RM, Scorza CA, Arida RM, Cavalheiro EA, de Almeida AC, Scorza FA. Disciplina de Neurologia Experimental, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brasil. To investigate whether n-3 polyunsaturated fatty acids (n-3 PUFAs) would promote different morphological changes in the hippocampal formation of rats with epilepsy, we performed an immunocytochemical study using parvalbumin (PV) and calretinin (CR) distribution as a marker. Animals subjected to the experimental model of epilepsy with a single dose of pilocarpine were randomly divided into the following groups: animals with epilepsy treated daily with vehicle (EV) and animals with epilepsy treated daily with 85 mg/kg n-3 PUFAs (EW). Control animals administered saline were also randomly divided into two other groups: animals treated daily with vehicle (CV) and animals treated daily with 85 mg/kg n-3 PUFAs (CW). A larger number of PV-positive neurons were observed in EW when compared with EV, CV, and CW. Similarly, there were significantly more CR-positive neurons in EW than in EV. These findings demonstrate that omega-3 fatty acids prevent status epilepticus-associated neuropathological changes in the hippocampal formation of rats with epilepsy. PMID: 18295545 [PubMed - indexed for MEDLINE] 51. Neurol Sci. 2008 Jun;29(3):147-52. doi: 10.1007/s10072-008-0926-1. Epub 2008 Jul 9. The protective effect of fish n-3 fatty acids on cerebral ischemia in rat prefrontal cortex. Ozen OA, Cosar M, Sahin O, Fidan H, Eser O, Mollaoglu H, Alkoc O, Yaman M, Songur A. Kocatepe University, School of Medicine, Department of Anatomy, Afyonkarahisar, Turkey. This study presents neuroprotective effects of fish n-3 EFA on the prefrontal cortex after cerebral ischemia and reperfusion. Eighteen rats divided into three groups. Group A rats were used as control. Cerebral ischemia and reperfusion was produced in rats either on a standard diet (Group B) or a standard diet plus fish n-3 EFA for 14 days (Group C). The malondialdehyde (MDA) levels and activities of superoxide dismutase (SOD) and catalase (CAT) were measured and the number of apoptotic neurons was counted. The levels of MDA and activities of SOD increased in Group B rats as compared to Group A rats, and decreased in Group C rats as compared to Group B rats. The activities of CAT increased in Group C as compared to Group B rats. The number of apoptotic neurons in the prefrontal cortex was lower in Group C as compared to Group B rats. PMID: 18612761 [PubMed - indexed for MEDLINE] 52. Alzheimers Dement. 2008 Jan;4(1 Suppl 1):S153-68. doi: 10.1016/j.jalz.2007.10.005. Epub 2007 Dec 21. Oral administration of circulating precursors for membrane phosphatides can promote the synthesis of new brain synapses. Cansev M, Wurtman RJ, Sakamoto T, Ulus IH. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA. Although cognitive performance in humans and experimental animals can be improved by administering omega-3 fatty acid docosahexaenoic acid (DHA), the neurochemical mechanisms underlying this effect remain uncertain. In general, nutrients or drugs that modify brain function or behavior do so by affecting synaptic transmission, usually by changing the quantities of particular neurotransmitters present within synaptic clefts or by acting directly on neurotransmitter receptors or signal-transduction molecules. We find that DHA also affects synaptic transmission in mammalian brain. Brain cells of gerbils or rats receiving this fatty acid manifest increased levels of phosphatides and of specific presynaptic or postsynaptic proteins. They also exhibit increased numbers of dendritic spines on postsynaptic neurons. These actions are markedly enhanced in animals that have also received the other two circulating precursors for phosphatidylcholine, uridine (which gives rise to brain uridine diphosphate and cytidine triphosphate) and choline (which gives rise to phosphocholine). The actions of DHA aere reproduced by eicosapentaenoic acid, another omega-3 compound, but not by omega-6 fatty acid arachidonic acid. Administration of circulating phosphatide precursors can also increase neurotransmitter release (acetylcholine, dopamine) and affect animal behavior. Conceivably, this treatment might have use in patients with the synaptic loss that characterizes Alzheimer's disease or other neurodegenerative diseases or occurs after stroke or brain injury. PMCID: PMC2344157 PMID: 18631994 [PubMed - indexed for MEDLINE] 53. Neuromolecular Med. 2008;10(4):219-35. doi: 10.1007/s12017-008-8036-z. Epub 2008 Jun 10. Neurological benefits of omega-3 fatty acids. Dyall SC, Michael-Titus AT. British College of Osteopathic Medicine, Lief House, 120-122 Finchley Road, NW5 5HR, London, UK. sdyall@bcom.ac.uk The central nervous system is highly enriched in long-chain polyunsaturated fatty acid (PUFA) of the omega-6 and omega-3 series. The presence of these fatty acids as structural components of neuronal membranes influences cellular function both directly, through effects on membrane properties, and also by acting as a precursor pool for lipid-derived messengers. An adequate intake of omega-3 PUFA is essential for optimal visual function and neural development. Furthermore, there is increasing evidence that increased intake of the long-chain omega-3 PUFA, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), may confer benefits in a variety of psychiatric and neurological disorders, and in particular neurodegenerative conditions. However, the mechanisms underlying these beneficial effects are still poorly understood. Recent evidence also indicates that in addition to the positive effects seen in chronic neurodegenerative conditions, omega-3 PUFA may also have significant neuroprotective potential in acute neurological injury. Thus, these compounds offer an intriguing prospect as potentially new therapeutic approaches in both chronic and acute conditions. The purpose of this article is to review the current evidence of the neurological benefits of omega-3 PUFA, looking specifically at neurodegenerative conditions and acute neurological injury. PMID: 18543124 [PubMed - indexed for MEDLINE] 54. Brain Res Rev. 2007 Dec;56(2):443-71. Epub 2007 Oct 23. Comparison of biochemical effects of statins and fish oil in brain: the battle of the titans. Farooqui AA, Ong WY, Horrocks LA, Chen P, Farooqui T. Department of Molecular and Cellular Biochemistry, The Ohio State University, 1645 Neil Avenue, Columbus, OH 43210, USA. farooqui.1@osu.edu Neural membranes are composed of glycerophospholipids, sphingolipids, cholesterol and proteins. The distribution of these lipids within the neural membrane is not random but organized. Neural membranes contain lipid rafts or microdomains that are enriched in sphingolipids and cholesterol. These rafts act as platforms for the generation of glycerophospholipid-, sphingolipid-, and cholesterol-derived second messengers, lipid mediators that are necessary for normal cellular function. Glycerophospholipid-derived lipid mediators include eicosanoids, docosanoids, lipoxins, and platelet-activating factor. Sphingolipid-derived lipid mediators include ceramides, ceramide 1-phosphates, and sphingosine 1-phosphate. Cholesterol-derived lipid mediators include 24-hydroxycholesterol, 25-hydroxycholesterol, and 7-ketocholesterol. Abnormal signal transduction processes and enhanced production of lipid mediators cause oxidative stress and inflammation. These processes are closely associated with the pathogenesis of acute neural trauma (stroke, spinal cord injury, and head injury) and neurodegenerative diseases such as Alzheimer disease. Statins, the HMG-CoA reductase inhibitors, are effective lipid lowering agents that significantly reduce risk for cardiovascular and cerebrovascular diseases. Beneficial effects of statins in neurological diseases are due to their anti-excitotoxic, antioxidant, and anti-inflammatory properties. Fish oil omega-3 fatty acids, eicosapentaenoic acid and docosahexaenoic acid, have similar anti-excitotoxic, antioxidant and anti-inflammatory effects in brain tissue. Thus the lipid mediators, resolvins, protectins, and neuroprotectins, derived from eicosapentaenoic acid and docosahexaenoic acid retard neuroinflammation, oxidative stress, and apoptotic cell death in brain tissue. Like statins, ingredients of fish oil inhibit generation of beta-amyloid and provide protection from oxidative stress and inflammatory processes. Collective evidence suggests that antioxidant, anti-inflammatory, and anti-apoptotic properties of statins and fish oil contribute to the clinical efficacy of treating neurological disorders with statins and fish oil. We speculate that there is an overlap between neurochemical events associated with neural cell injury in stroke and neurodegenerative diseases. This commentary compares the neurochemical effects of statins with those of fish oil. PMID: 17959252 [PubMed - indexed for MEDLINE] 55. Brain. 2007 Nov;130(Pt 11):3004-19. Epub 2007 Sep 27. A combination of intravenous and dietary docosahexaenoic acid significantly improves outcome after spinal cord injury. Huang WL, King VR, Curran OE, Dyall SC, Ward RE, Lal N, Priestley JV, Michael-Titus AT. Neuroscience Centre, Institute of Cell & Molecular Science, Queen Mary University of London, UK. w.huang@qmul.ac.uk Previous studies have shown that omega-3 polyunsaturated fatty acids such as alpha-linolenic acid and docosahexaenoic acid (DHA) are neuroprotective in models of spinal cord injury (SCI) in rodents. However, the mechanism of action underlying these effects has not been elucidated, and the optimum treatment regime remains to be defined. We have therefore carried out a detailed analysis of the effects of DHA in adult rats subject to thoracic compression SCI. Saline or DHA (250 nmol/kg) was administered intravenously (i.v.) 30 min after compression. After injury, the saline group received a standard control diet for 1 or 6 weeks, whereas DHA-injected animals received either a control or a DHA-enriched diet (400 mg/kg/day) for 1 or 6 weeks. Other groups received a DHA-enriched diet only for 1 week following injury, or received acute DHA (250 nmol/kg; i.v.) treatment delayed up to 3 h after injury. We also assessed oxidative stress and the inflammatory reaction at the injury site, neuronal and oligodendrocyte survival and axonal damage and the locomotor recovery. At 24 h, lipid peroxidation, protein oxidation, RNA/DNA oxidation and the induction of cyclooxygenase-2 were all significantly reduced by i.v. DHA administration. At 1 week and 6 weeks, macrophage recruitment was reduced and neuronal and oligodendrocyte survival was substantially increased. Axonal injury was reduced at 6 weeks. Locomotor recovery was improved from day 4, and sustained up to 6 weeks. Rats treated with a DHA-enriched diet in addition to the acute DHA injection were not significantly different from the acute DHA-treated animals at 1 week, but at 6 weeks showed additional improvements in both functional and histological outcomes. DHA treatment was ineffective if the acute injection was delayed until 3 h post-injury, or if the DHA was administered for 1 week solely by diet. Our results in a clinically relevant model of SCI show that significant neuroprotection can be obtained by combining an initial acute i.v. injection of DHA with a sustained dietary supplementation. Given that the safety and tolerability of preparations enriched in omega-3 fatty acids is already well-documented, such a combined DHA treatment regime deserves consideration as a very promising approach to SCI management. PMID: 17901087 [PubMed - indexed for MEDLINE] 56. Neurotoxicology. 2007 Nov;28(6):1220-9. Epub 2007 Aug 10. Detrimental effects of post-treatment with fatty acids on brain injury in ischemic rats. Yang DY, Pan HC, Yen YJ, Wang CC, Chuang YH, Chen SY, Lin SY, Liao SL, Raung SL, Wu CW, Chou MC, Chiang AN, Chen CJ. Chang Bing Show Chwan Memorial Hospital, Changhua 542, Taiwan. Studies have illustrated that fatty acids, especially polyunsaturated fatty acids (PUFA), have a role in regulating oxidative stress via the enhancement of antioxidative defense capacity or the augmentation of oxidative burden. Elevated oxidative stress has been implicated in the pathogenesis of brain injury associated with cerebral ischemia/reperfusion (I/R). The objective of this study was to assess whether treatment with fatty acids after focal cerebral I/R induced by occlusion of the common carotid arteries and the middle cerebral artery has effects on brain injury in a rat model. PUFA, including arachidonic acid (AA) and docosahexaenoic acid (DHA), and the saturated fatty acid, stearic acid (SA), were administrated 60 min after reperfusion via intraperitoneal injection. AA and DHA aggravated cerebral ischemic injury, which manifested as enlargement of areas of cerebral infarction and increased impairment of motor activity, in a concentration-dependent manner. However, there were no remarkable differences in post-ischemic alterations between the SA and saline groups. The post-ischemic augmentation of injury in AA and DHA treatment groups was accompanied by increases in the permeability of the blood-brain barrier (BBB), brain edema, metalloproteinase (MMP) activity, inflammatory cell infiltration, cyclooxygenase 2 (COX-2) expression, caspase 3 activity, and malondialdehyde (MDA) production, and by a decrease in the brain glutathione (GSH) content. Furthermore, we found that either AA or DHA alone had little effect on free radical generation in neuroglia, but they greatly increased the hydrogen peroxide-induced oxidative burden. Taken together, these findings demonstrate the detrimental effect of PUFA such as AA and DHA in post-ischemic progression and brain injury after cerebral I/R is associated with augmentation of cerebral I/R-induced alterations, including oxidative changes. PMID: 17854901 [PubMed - indexed for MEDLINE] 57. J Neurotrauma. 2007 Oct;24(10):1587-95. Omega-3 fatty acids supplementation restores mechanisms that maintain brain homeostasis in traumatic brain injury. Wu A, Ying Z, Gomez-Pinilla F. Department of Physiological Science, University of California at Los Angeles (UCLA), Los Angeles, California 90095, USA. Traumatic brain injury (TBI) produces a state of vulnerability that reduces the brain capacity to cope with secondary insults. The silent information regulator 2 (Sir2) has been implicated with maintaining genomic stability and cellular homeostasis under challenging situation. Here we explore the possibility that the action of Sir2alpha (mammalian Sir2) in the brain can extend to serve neuronal plasticity. We provide novel evidence showing that mild TBI reduces the expression of Sir2alpha in the hippocampus, in proportion to increased levels of protein oxidation. In addition, we show that dietary supplementation of omega-3 fatty acids that ameliorates protein oxidation was effective to reverse the reduction of Sir2alpha level in injured rats. Given that oxidative stress is a subproduct of dysfunctional energy homeostasis, we measured AMP-activated protein kinase (AMPK) and phosphorylated-AMPK (p-AMPK) to have an indication of the energy status of cells. Hippocampal levels of total and phosphorylated AMPK were reduced after TBI and levels were normalized by omega-3 fatty acts supplements. Further, we found that TBI reduced ubiquitous mitochondrial creatine kinase (uMtCK), an enzyme implicated in the energetic regulation of Ca2+-pumps and in the maintenance of Ca2+-homeostasis. Omega-3 fatty acids supplements normalized the levels of uMtCK after lesion. Furthermore, we found that the correlation between Sir2alpha and AMPK or p-AMPK was disrupted by TBI, but restored by omega-3 fatty acids supplements. Our results suggest that TBI may compromise neuronal protective mechanisms by involving the action of Sir2alpha. In addition, results show the capacity of omega-3 fatty acids to counteract some of the effects of TBI by normalizing levels of molecular systems associated with energy homeostasis. PMID: 17970622 [PubMed - indexed for MEDLINE] 58. Neurochem Int. 2007 Feb;50(3):548-54. Epub 2006 Dec 21. The protective effect of fish n-3 fatty acids on cerebral ischemia in rat hippocampus. Bas O, Songur A, Sahin O, Mollaoglu H, Ozen OA, Yaman M, Eser O, Fidan H, Yagmurca M. Kocatepe University, School of Medicine, Department of Anatomy, Afyonkarahisar, Turkey. orhanbas55@hotmail.com Reactive oxygen species (ROS) have been implicated in the pathogenesis of cerebral injury after ischemia-reperfusion (I/R). Fish n-3 essential fatty acids (EFA), contain eicosapentaenoic acids (EPA) and docosahexoenoic acids (DHA), exhibit antioxidant properties. DHA is an important component of brain membrane phospholipids and is necessary for the continuity of neuronal functions. EPA prevents platelet aggregation and inhibits the conversion of arachidonic acid into thromboxane A(2) and prostaglandins. They have been suggested to be protective agents against neurological and neuropsychiatric disorders. In this study, the neuroprotective effects of fish n-3 EFA on oxidant-antioxidant systems and number of apoptotic neurons of the hippocampal formation (HF) subjected to cerebral I/R injury was investigated in Sprague-Dawley rats. Six rats were used as control (Group I). Cerebral ischemia was produced by occlusion of both the common carotid arteries combined with hypotension for 45 min, followed by reperfusion for 30 min, in rats either on a standard diet (Group II) or a standard diet plus fish n-3 EFA (Marincap((R)), 0.4 g/kg/day, by gavage) for 14 days (Group III). At the end of procedures, the rats were sacrificed and their brains were removed immediately. The levels of malonedialdehyde (MDA) and nitric oxide (NO) and activities of superoxide dismutase (SOD) and catalase (CAT) were measured in left HF. In addition, the number of apoptotic neurons was counted by terminal transferase dUTP nick end labelling (TUNEL) assay in histological samples of the right HF. We found that SOD activities and MDA levels increased in Group III rats compared with Group II rats. On the other hand, CAT activities and NO levels were found to be decreased in Group III rats compared with Group II rats. Additionally, the number of apoptotic neurons was lower in Group III in comparison with Group II rats. The present findings suggest that fish n-3 EFA could decrease the oxidative status and apoptotic changes in ischemic rat hippocampal formation. Dietary supplementation of n-3 EFA may be beneficial to preserve or ameliorate ischemic cerebral vascular disease. PMID: 17187901 [PubMed - indexed for MEDLINE] 59. J Am Soc Mass Spectrom. 2007 Jan;18(1):128-44. Epub 2006 Oct 19. Resolvin D1, protectin D1, and related docosahexaenoic acid-derived products: Analysis via electrospray/low energy tandem mass spectrometry based on spectra and fragmentation mechanisms. Hong S, Lu Y, Yang R, Gotlinger KH, Petasis NA, Serhan CN. Analytical Core, Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA. Resolvin D1 (RvD1) and protectin D1 (Neuroprotectin D1, PD1/NPD1) are newly identified anti-inflammatory lipid mediators biosynthesized from docosahexaenoic acid (DHA). In this report, the spectra-structure correlations and fragmentation mechanisms were studied using electrospray low-energy collision-induced dissociation tandem mass spectrometry (MS/MS) for biogenic RvD1 and PD1, as well as mono-hydroxy-DHA and related hydroperoxy-DHA. The loss of H2O and CO2 in the spectra indicates the number of functional group(s). Chain-cut ions are the signature of the positions and numbers of functional groups and double bonds. The observed chain-cut ion is equivalent to a hypothetical homolytic-segment (cc, cm, mc, or mm) with addition or extraction of up to 2 protons (H). The alpha-cleavage ions are equivalent to: [cc + H], with H from the hydroxyl through a beta-ene or gamma-ene rearrangement; [cm - 2H], with 2H from hydroxyls of PD1 through a gamma-ene rearrangement, or 1H from the hydroxyl and the other H from the alpha-carbon of mono-HDHA through an alpha-H-beta-ene rearrangement; [mc - H], with H from hydroxyl through a beta-ene or gamma-ene rearrangement, or from the alpha-carbon through an alpha-H-beta-ene rearrangement; or [mm] through charge-direct fragmentations. The beta-ene or gamma-ene facilitates the H shift to gamma position and alpha-cleavage. Deuterium labeling confirmed the assignment of MS/MS ions and the fragmentation mechanisms. Based on the MS/MS spectra and fragmentation mechanisms, we identified RvD1, PD1, and mono-hydroxy-DHA products in human neutrophils and blood, trout head-kidney, and stroke-injury murine brain-tissue. PMCID: PMC2763184 PMID: 17055291 [PubMed - indexed for MEDLINE] 60. Methods Enzymol. 2007;433:127-43. Measurement of products of docosahexaenoic acid peroxidation, neuroprostanes, and neurofurans. Arneson KO, Roberts LJ 2nd. Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA. Free radicals derived primarily from oxygen have been implicated in the pathophysiology of a wide variety of human diseases. Quantification of products of free radical damage in biological systems is necessary to understand the role of free radicals in disease states. Measures of lipid peroxidation are often used to quantitate oxidative damage though many of these measures have inherent problems with sensitivity and specificity especially when used to quantitate in vivo oxidative injury. The discovery of the F(2)-isoprostanes (F(2)-IsoPs), prostaglandin F(2)-like compounds derived by the free radical peroxidation of arachidonic acid (AA, C20:4, omega-6) has largely overcome these limitations. The measurement of the F(2)-IsoPs has been shown to be one of the most accurate approaches to quantifying oxidative damage in vivo. We have extended our studies of lipid peroxidation and the F(2)-IsoPs to docosahexaenoic acid (DHA, C22:6, omega-3) and its peroxidation products. We have found that DHA oxidizes both in vitro and in vivo to form F(2)-IsoP-like compounds termed F(4)-neuroprostanes (F(4)-NPs). DHA is specifically enriched in neuronal membranes making the F(4)-NPs sensitive and specific markers of neuronal oxidative damage. Adapting the methodology used to quantitate the F(2)-IsoPs, we utilize stable isotope dilution, negative ion chemical ionization, gas chromatography mass spectrometry (GC/MS) to quantitate the F(4)-NPs with a limit of detection in the low picomolar range. Methods have been developed to quantitate both the F(4)-NPs and the neurofurans (NFs), DHA derived peroxidation products containing a substituted tetrahydrofuran ring, in brain tissue and cerebrospinal fluid. This review outlines in detail proper sample handling, extraction and hydrolysis of the F(4)-NPs and NFs from tissue membrane phospholipids or biological fluids, and purification and derivatization of the compounds for analysis by GC/MS. PMID: 17954232 [PubMed - indexed for MEDLINE] 61. Neurosci Res. 2006 Oct;56(2):159-64. Epub 2006 Aug 14. Dietary supplementation of arachidonic and docosahexaenoic acids improves cognitive dysfunction. Kotani S, Sakaguchi E, Warashina S, Matsukawa N, Ishikura Y, Kiso Y, Sakakibara M, Yoshimoto T, Guo J, Yamashima T. Department of Neurosurgery, Minami-gaoka Hospital, Kanazawa, Ishikawa, Japan. Age-dependent increase of peroxidation of membrane fatty acids such as arachidonic acid (ARA) and docosahexaenoic acid (DHA) in neurons was reported to cause a decline of the hippocampal long-term potentiation (LTP) and cognitive dysfunction in rodents. Although supplementation of ARA and DHA can improve LTP and cognitive function in rodents, their effects in humans are unknown. The present work was undertaken to study whether ARA and DHA have beneficial effects in human amnesic patients. The subjects were 21 mild cognitive dysfunction (12 MCI-A with supplementation and 9 MIC-P with placebo), 10 organic brain lesions (organic), and 8 Alzheimer's disease (AD). The cognitive functions were evaluated using Japanese version of repeatable battery for assessment of neuropsychological status (RBANS) at two time points: before and 90 days after the supplementation of 240 mg/day ARA and DHA, or 240 mg/day of olive oil, respectively. MCI-A group showed a significant improvement of the immediate memory and attention score. In addition, organic group showed a significant improvement of immediate and delayed memories. However, there were no significant improvements of each score in AD and MCI-P groups. It is suggested from these data that ARA and DHA supplementation can improve the cognitive dysfunction due to organic brain damages or aging. PMID: 16905216 [PubMed - indexed for MEDLINE] 62. Cell Mol Neurobiol. 2006 Jul-Aug;26(4-6):901-13. Epub 2006 Aug 1. The onset of brain injury and neurodegeneration triggers the synthesis of docosanoid neuroprotective signaling. Bazan NG. LSU Neuroscience Center and Department of Ophthalmology, Louisiana State University Health Sciences Center School of Medicine in New Orleans, New Orleans 70112, USA. nbazan@lsuhsc.edu Bioactive lipid messengers are formed through phospholipase-mediated cleavage of specific phospholipids from membrane reservoirs. Effectors that activate the synthesis of lipid messengers, include ion channels, neurotransmitters, membrane depolarization, cytokines, and neurotrophic factors. In turn, lipid messengers regulate and interact with multiple pathways, participating in the development, differentiation, function (e.g., long-term potentiation and memory), protection, and repair of cells of the nervous system. Overall, bioactive lipids participate in the regulation of synaptic function and dysfunction. Platelet-activating factor (PAF) and COX-2-synthesized PGE(2) modulate synaptic plasticity and memory. Oxidative stress disrupts lipid signaling, fosters lipid peroxidation, and initiates and propagates neurodegeneration. Lipid messengers participate in the interactions among neurons, astrocytes, oligodendrocytes, microglia, cells of the microvasculature, and other cells. A conglomerate of interrelated cells comprises the neurovascular unit. Signaling at the neurovascular unit is clearly altered in the early stages of cerebrovascular disease as well as in neurodegenerations. Here we will provide examples of how signaling by lipids regulates critical events essential for neuronal survival. We will highlight a newly identified, DHA-derived messenger, neuroprotectin D1, which attenuates oxidative stress-induced apoptosis. The specificity and potency of this novel docosanoid (neuroprotectin D1) indicate a potentially important target for therapeutic intervention. PMID: 16897369 [PubMed - indexed for MEDLINE] 63. Neuroscience. 2006;137(1):241-51. Epub 2005 Nov 14. Alpha-linolenic acid and riluzole treatment confer cerebral protection and improve survival after focal brain ischemia. Heurteaux C, Laigle C, Blondeau N, Jarretou G, Lazdunski M. Institut de Pharmacologie Moléculaire et Cellulaire, UMR 6097, CNRS Université de Nice Sophia Antipolis, Institut Paul Hamel, 660 Route des Lucioles, Sophia-Antipolis, 06560 Valbonne, France. heurteau@ipmc.cnrs.fr We investigated here the effects of alpha-linolenic acid and riluzole, both activators of the 2P-domain K+ channel family TREK/TRAAK, in a model of focal ischemia clinically relevant to stroke, not only assessing neuronal protection, but also long term survival. Moreover, all the drug treatments were initiated post-ischemia. Mice were subjected to transient middle cerebral artery occlusion (1 h) and reperfusion according to the intraluminal filament model. Drugs were injected into the jugular vein according to three protocols: (i) a single dose of 4 mg/kg riluzole or 500 nmol/kg alpha-linolenic acid at different reperfusion time; (ii) a three-day therapy (a single dose of 2 mg/kg riluzole and 250 nmol/kg alpha-linolenic acid given 1-2, 48 and 72 h after reperfusion); (iii) a three-week therapy (a single dose of 2 mg/kg riluzole and 250 nmol/kg alpha-linolenic acid given once a week during three weeks after reperfusion. A combined treatment with 2mg/kg riluzole+250 nmol/kg alpha-linolenic acid injected 2 h after reperfusion was also tested. A single dose of riluzole (4 mg/kg) or alpha-linolenic acid (500 nmol/kg) injected up to 3 h after reperfusion reduced drastically the stroke volume by 75% and 86%, respectively. Neurological deficits 24 h after ischemia were significantly improved by alpha-linolenic acid500 or riluzole4 with a neurological score of 1.8 as compared with 2.5 observed in vehicle-treated mice. Alpha-linolenic acid- and riluzole treatment were associated with a reduction in cytopathological features of cell injury, including DNA fragmentation and Bax expression in the cortex and the caudate putamen. With regard to the survival rate at 30 days, the best protections were obtained with the alpha-linolenic acid-injection in the three-week therapy as well as with a single dose of the combined treatment (2 mg/kg riluzole+250 nmol/kg alpha-linolenic acid). Palmitic acid, a saturated fatty acid that does not activate the 2P-domain K-channel TREK/TRAAK family, did not provide any neuroprotection. Taken together, these data suggest that the TREK/TRAAK K-channel family may be a promising target for neuroprotection, and that riluzole and alpha-linolenic acid could be of therapeutic value against focal ischemia/reperfusion injury to the brain. PMID: 16289892 [PubMed - indexed for MEDLINE] 64. Nutr Health. 2006;18(3):277-84. The impact of diet and exercise on brain plasticity and disease. Pinilla FG. Department of Neurosurgery, Brain Injury Research Center, UCLA School of Medicine, Los Angeles, California 90095, USA. Fgomezpi@ucla.ed Lifestyle involves our preference to engage in behaviors that can remarkably influence the fitness level of our body and brain. Dietary factors are a powerful means to influence brain function on a daily basis. We have shown that the consumption of a diet rich in saturated fat decreases learning and memory and increases metabolic distress. Conversely, diets supplemented either with omega-3 fatty acids, vitamin E or the curry spice curcumin benefit cognitive function. Equally impressive is the action of exercise on cognitive function as documented by studies showing that exercise enhances learning and memory. The beneficial action of exercise on the brain can be used therapeutically to overcome the effects of consuming a poor diet. We suggest that the managed use of diet and exercise can help the brain to cope with several types of insults and ultimately benefit brain function. PMID: 17180873 [PubMed - indexed for MEDLINE] 65. J Biol Chem. 2005 Dec 30;280(52):43079-86. Epub 2005 Oct 10. The docosatriene protectin D1 is produced by TH2 skewing and promotes human T cell apoptosis via lipid raft clustering. Ariel A, Li PL, Wang W, Tang WX, Fredman G, Hong S, Gotlinger KH, Serhan CN. Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA. Docosahexaenoic acid, a major omega-3 fatty acid in human brain, synapses, retina, and other neural tissues, displays beneficial actions in neuronal development, cancer, and inflammatory diseases by mechanisms that remain to be elucidated. In this study we found, using lipid mediator informatics employing liquid chromatography-tandem mass spectrometry, that (10,17S)-docosatriene/neuroprotectin D1, now termed protectin D1 (PD1), is generated from docosahexaenoic acid by T helper type 2-skewed peripheral blood mononuclear cells in a lipoxygenase-dependent manner. PD1 blocked T cell migration in vivo, inhibited tumor necrosis factor alpha and interferon-gamma secretion, and promoted apoptosis mediated by raft clustering. These results demonstrated novel anti-inflammatory roles for PD1 in regulating events associated with inflammation and resolution. PMID: 16216871 [PubMed - indexed for MEDLINE] 66. Ann N Y Acad Sci. 2005 Aug;1053:137-47. Brain response to injury and neurodegeneration: endogenous neuroprotective signaling. Bazan NG, Marcheselli VL, Cole-Edwards K. LSU Neuroscience Center and Department of Ophthalmology, Louisiana State University Health Sciences Center School of Medicine, 2020 Gravier Street, Suite D, New Orleans, Louisiana 70112, USA. nbazan@lsuhsc.edu Synaptic activity and ischemia/injury promote lipid messenger formation through phospholipase-mediated cleavage of specific phospholipids from membrane reservoirs. Lipid messengers modulate signaling cascades, contributing to development, differentiation, function (e.g., memory), protection, regeneration, and repair of neurons and overall regulation of neuronal, glial, and endothelial cell functional integrity. Oxidative stress disrupts lipid signaling and promotes lipid peroxidation and neurodegeneration. Lipid signaling at the neurovascular unit (neurons, astrocytes, oligodendrocytes, microglia, and cells of the microvasculature) is altered in early cerebrovascular and neurodegenerative disease. We discuss how lipid signaling regulates critical events in neuronal survival. Aberrant synaptic plasticity (e.g., epileptogenesis) is highlighted to show how gene expression may drive synaptic circuitry formation in the "wrong" direction. Docosahexaenoic acid has been implicated in memory, photoreceptor cell biogenesis and function, and neuroprotection. Free docosahexaenoic acid released in the brain during experimental stroke leads to the synthesis of stereospecific messengers through oxygenation pathways. One messenger, 10,17S-docosatriene (neuroprotectin D1; NPD1), counteracts leukocyte infiltration and proinflammatory gene expression in brain ischemia-reperfusion. In retina, photoreceptor survival depends on retinal pigment epithelial (RPE) cell integrity. NPD1 is synthesized in RPE cells undergoing oxidative stress, potently counteracts oxidative stress-triggered apoptotic DNA damage in RPE, upregulates antiapoptotic proteins Bcl-2 and Bcl-x(L), and decreases proapoptotic Bax and Bad expression. These findings expand our understanding of how the nervous system counteracts redox disturbances, mitochondrial dysfunction, and proinflammatory conditions. The specificity and potency of NPD1 indicate a potential target for therapeutic intervention for stroke, age-related macular degeneration, spinal cord injury, and other neuroinflammatory or neurodegenerative diseases. PMID: 16179516 [PubMed - indexed for MEDLINE] 67. Brain Pathol. 2005 Apr;15(2):159-66. Neuroprotectin D1 (NPD1): a DHA-derived mediator that protects brain and retina against cell injury-induced oxidative stress. Bazan NG. LSU Neuroscience Center and Department of Opthamology, Louisiana State University Health Sciences Center School of Medicine, New Orleans 70112, USA. nbazan@lsuhsc.edu The biosynthesis of oxygenated arachidonic acid messengers triggered by cerebral ischemia-reperfusion is preceded by an early and rapid phospholipase A2 activation reflected in free arachidonic and docosahexaenoic acid (DHA) accumulation. These fatty acids are released from membrane phospholipids. Both fatty acids are derived from dietary essential fatty acids; however, only DHA, the omega-3 polyunsaturated fatty acyl chain, is concentrated in phospholipids of various cells of brain and retina. Synaptic membranes and photoreceptors share the highest content of DHA of all cell membranes. DHA is involved in memory formation, excitable membrane function, photoreceptor cell biogenesis and function, and neuronal signaling, and has been implicated in neuroprotection. In addition, this fatty acid is required for retinal pigment epithelium cell (RPE) functional integrity. Here we provide an overview of the recent elucidation of a specific mediator generated from DHA that contributes at least in part to its biological significance. In oxidative stress-challenged human RPE cells and rat brain undergoing ischemia-reperfusion, 10,17S-docosatriene (neuroprotectin D1, NPD1) synthesis evolves. In addition, calcium ionophore A23187, IL-1beta, or the supply of DHA enhances NPD1 synthesis. A time-dependent release of endogenous free DHA followed by NPD1 formation occurs, suggesting that a phospholipase A2 releases the mediator's precursor. When NPD1 is infused during ischemia-reperfusion or added to RPE cells during oxidative stress, apoptotic DNA damage is down-regulated. NPD1 also up-regulates the anti-apoptotic Bcl-2 proteins Bcl-2 and BclxL and decreases pro-apoptotic Bax and Bad expression. Moreover, NPD1 inhibits oxidative stress-induced caspase-3 activation. NPD1 also inhibits IL-1beta-stimulated expression of COX-2. Overall, NPD1 protects cells from oxidative stress-induced apoptosis. Because photoreceptors are progressively impaired after RPE cell damage in retinal degenerative diseases, understanding of how these signals contribute to retinal cell survival may lead to the development of new therapeutic strategies. Moreover, NPD1 bioactivity demonstrates that DHA is not only a target of lipid peroxidation, but rather is the precursor to a neuroprotective signaling response to ischemia-reperfusion, thus opening newer avenues of therapeutic exploration in stroke, neurotrauma, spinal cord injury, and neurodegenerative diseases, such as Alzheimer disease, aiming to up-regulate this novel cell-survival signaling. PMID: 15912889 [PubMed - indexed for MEDLINE] 68. Stroke. 2005 Jan;36(1):118-23. Epub 2004 Nov 29. Docosahexaenoic acid complexed to albumin elicits high-grade ischemic neuroprotection. Belayev L, Marcheselli VL, Khoutorova L, Rodriguez de Turco EB, Busto R, Ginsberg MD, Bazan NG. Cerebral Vascular Disease Research Center, Department of Neurology, University of Miami School of Medicine, Miami, FL 33101, USA. BACKGROUND AND PURPOSE: High-dose human albumin therapy is strongly neuroprotective in models of brain ischemia and trauma and is currently being studied in a pilot-phase clinical stroke trial. Among its actions in ischemia, albumin induces the systemic mobilization of n-3 polyunsaturated fatty acids and may help to replenish polyunsaturated fatty acids lost from neural membranes. METHODS: We complexed 25% human albumin to docosahexaenoic acid (DHA; 22:6n-3) and compared its neuroprotective efficacy with that of native albumin in rats with 2-hour focal ischemia produced by intraluminal suture-occlusion of the middle cerebral artery. RESULTS: In animals treated with DHA-albumin, 0.63 g/kg, the improvement in neurobehavioral scores at 72 hours significantly exceeded that of other treatment groups, and the extent of histological protection (86% reduction in cortical infarction) was highly significant and tended to surpass the degree of cortical protection produced by native albumin at 1.25 g/kg (65%). DHA-albumin 0.63 g/kg, but not native albumin, also significantly reduced subcortical infarction and markedly diminished brain swelling. Lipidomic analysis of DHA-albumin-treated postischemic brains revealed a large accumulation of the neuroprotective DHA metabolite, 10,17S-docosatriene, in the ipsilateral hemisphere. CONCLUSIONS: The high-grade neuroprotection afforded by the DHA-albumin complex at relatively low albumin doses is clinically advantageous in that it might reduce the likelihood of acute intravascular volume overload and congestive heart failure sometimes induced when patients with compromised cardiovascular function are treated with high-dose albumin. PMID: 15569878 [PubMed - indexed for MEDLINE] 69. Pharmacol Biochem Behav. 2004 Dec;79(4):651-9. Protective effect of chronic ethyl docosahexaenoate administration on brain injury in ischemic gerbils. Cao DH, Xu JF, Xue RH, Zheng WF, Liu ZL. Department of Biology, Nanjing University, 22 Hankou Road, Jiangsu 210093, PR China. There is evidence that the excessive generation of reactive oxygen free radicals contributes to the brain injury associated with cerebral ischemia. In the present study, the protective effect of chronic administration of ethyl docosahexaenoate (E-DHA) against oxidative brain injury was evaluated in the gerbil model of transient cerebral ischemia. Weanling male gerbils were orally pretreated with either E-DHA (200 mg/kg) or vehicle, once a day, for 10 weeks and subjected to bilateral occlusion of common carotid arteries for 10 min. At the different reperfusion times, E-DHA pretreatment significantly inhibited the increases in the production of brain salicylate-derived 2,5-dihydroxybenzoic acid (2,5-DHBA) and content of brain malonildialdehyde (MDA). The superoxide dismutase (SOD) activity was not modified; however, pretreatment with E-DHA significantly prevented the level of brain-reduced glutathione (GSH) and activities of brain glutathione peroxidase (GSH-P(X)) and catalase (CAT) from declines caused by cerebral ischemia. Moreover, ischemia and reperfusion-induced delayed neuronal loss in the hippocampus CA1 sector and locomotor hyperactivity were also significantly attenuated by pretreatment with E-DHA. These results suggested that the neuroprotective effect of E-DHA might be due to its antioxidant property. PMID: 15582673 [PubMed - indexed for MEDLINE] 70. J Neurochem. 2004 Oct;91(1):20-9. Neuroprotective actions of eicosapentaenoic acid on lipopolysaccharide-induced dysfunction in rat hippocampus. Lonergan PE, Martin DS, Horrobin DF, Lynch MA. Trinity College Institute of Neuroscience, Department of Physiology, Trinity College, Dublin, Ireland. Eicosapentaenoic acid (EPA) protects hippocampus from age-related and irradiation-induced changes that lead to impairment in synaptic function; the evidence suggests that this is due to its anti-inflammatory effects, specifically preventing changes induced by the proinflammatory cytokine, interleukin-1beta (IL-1beta). In this study, we have investigated the possibility that EPA may prevent the effects of lipopolysaccharide (LPS) administration, which have been shown to lead to deterioration of synaptic function in rat hippocampus. The data indicate that treatment of hippocampal neurones with EPA abrogated the LPS-induced increases in phosphorylation of the mitogen-activated protein kinase, c-Jun N-terminal kinase (JNK), the transcription factor, c-Jun and the mitochondrial protein, Bcl-2. In parallel, we report that intraperitoneal administration of LPS to adult rats increases phosphorylation of JNK, c-Jun and Bcl-2 in hippocampal tissue and that these changes are coupled with increased IL-1beta concentration. Treatment of rats with EPA abrogates these effects and also blocks the LPS-induced impairment in long-term potentiation in perforant path-granule cell synapses that accompanies these changes. We propose that the neuroprotective effect of EPA may be dependent on its ability to inhibit the downstream consequences of JNK activation. PMID: 15379883 [PubMed - indexed for MEDLINE] 71. J Neurotrauma. 2004 Oct;21(10):1457-67. Dietary omega-3 fatty acids normalize BDNF levels, reduce oxidative damage, and counteract learning disability after traumatic brain injury in rats. Wu A, Ying Z, Gomez-Pinilla F. Department of Physiological Science, University of California at Los Angeles, 90095, USA. Comment in J Neurotrauma. 2008 Dec;25(12):1499. Omega-3 fatty acids (i.e., docosahexaenoic acid; DHA) regulate signal transduction and gene expression, and protect neurons from death. In this study we examined the capacity of dietary omega3 fatty acids supplementation to help the brain to cope with the effects of traumatic injury. Rats were fed a regular diet or an experimental diet supplemented with omega-3 fatty acids, for 4 weeks before a mild fluid percussion injury (FPI) was performed. FPI increased oxidative stress, and impaired learning ability in the Morris water maze. This type of lesion also reduced levels of brain-derived neurotrophic factor (BDNF), synapsin I, and cAMP responsive element-binding protein (CREB). It is known that BDNF facilitates synaptic transmission and learning ability by modulating synapsin I and CREB. Supplementation of omega-3 fatty acids in the diet counteracted all of the studied effects of FPI, that is, normalized levels of BDNF and associated synapsin I and CREB, reduced oxidative damage, and counteracted learning disability. The reduction of oxidative stress indicates a benevolent effect of this diet on mechanisms that maintain neuronal function and plasticity. These results imply that omega-3 enriched dietary supplements can provide protection against reduced plasticity and impaired learning ability after traumatic brain injury. PMID: 15672635 [PubMed - indexed for MEDLINE] 72. J Neurochem. 2003 Dec;87(6):1518-26. Pharmacologic suppression of neuronal oxidative damage and dendritic degeneration following direct activation of glial innate immunity in mouse cerebrum. Milatovic D, Zaja-Milatovic S, Montine KS, Horner PJ, Montine TJ. Department of Pathology, University of Washington, Harborview Medical Center, Seattle, Washington 98104, USA. Activation of glial innate immunity is widely proposed to contribute to a number of degenerative and destructive diseases of brain. However, the precise role of activated innate immunity has been difficult to define in vivo because of multiple simultaneous pathogenic processes and responses to injury that confound interpretation of results from complex models of disease. Here, we used the model of intracerebroventricular (ICV) injection of lipopolysaccharide (LPS) to test the hypothesis that directly activated glial innate immunity leads to neurodegeneration in cerebrum and to establish the molecular determinants of and neuroprotectants from such innate immunity-mediated neuronal damage. Our results showed that ICV LPS induced delayed, reversible oxidative damage to cerebral neuronal membranes as measured by F4-neuroprostanes that was coincident with degeneration of the hippocampal pyramidal neuron dendritic system, but not neuron death, in adult mice. Both neuronal oxidative damage and dendritic degeneration were NF-kappaB and iNOS dependent and were completely suppressed by ibuprofen and alpha-tocopherol, but not naproxen or gamma-tocopherol. These results prove that activation of glial innate immunity can lead to neurodegeneration independent of other pathologic processes, closely associate oxidative damage to neuronal membranes with degeneration of the dendritic system, and provide a possible explanation for the varying efficacy of neuroprotectants that have been suggested in observational studies of dementia. PMID: 14713307 [PubMed - indexed for MEDLINE] 73. J Biol Chem. 2003 Oct 31;278(44):43807-17. Epub 2003 Aug 15. Novel docosanoids inhibit brain ischemia-reperfusion-mediated leukocyte infiltration and pro-inflammatory gene expression. Marcheselli VL, Hong S, Lukiw WJ, Tian XH, Gronert K, Musto A, Hardy M, Gimenez JM, Chiang N, Serhan CN, Bazan NG. Neuroscience Center of Excellence and Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA. Erratum in J Biol Chem. 2003 Dec 19;278(51):51974. Ischemic stroke triggers lipid peroxidation and neuronal injury. Docosahexaenoic acid released from membrane phospholipids during brain ischemia is a major source of lipid peroxides. Leukocyte infiltration and pro-inflammatory gene expression also contribute to stroke damage. In this study using lipidomic analysis, we have identified stereospecific messengers from docosahexaenoate-oxygenation pathways in a mouse stroke model. Aspirin, widely used to prevent cerebrovascular disease, activates an additional pathway, which includes the 17R-resolvins. The newly discovered brain messenger 10,17S-docosatriene potently inhibited leukocyte infiltration, NFkappaB, and cyclooxygenase-2 induction in experimental stroke and elicited neuroprotection. In addition, in neural cells in culture, this lipid messenger also inhibited both interleukin 1-beta-induced NFkappaB activation and cyclooxygenase-2 expression. Thus, the specific novel bioactive docosanoids generated in vivo counteract leukocyte-mediated injury as well as pro-inflammatory gene induction. These results challenge the view that docosahexaenoate only participates in brain damage and demonstrate that this fatty acid is also the endogenous precursor to a neuroprotective signaling response to ischemia-reperfusion. PMID: 12923200 [PubMed - indexed for MEDLINE] 74. Neurosci Lett. 2003 Oct 2;349(2):136-8. Free fatty acids in cerebrospinal fluids from patients with traumatic brain injury. Pilitsis JG, Coplin WM, O'Regan MH, Wellwood JM, Diaz FG, Fairfax MR, Michael DB, Phillis JW. Department of Neurosurgery, Wayne State University School of Medicine, UHC-6E, 4201 St. Antoine, Detroit, MI 48201, USA. Free fatty acid (FFA) concentrations in cerebrospinal fluid (CSF) are recognized as markers of brain damage in animal studies. There is, however, relatively little information regarding FFA concentrations in human CSF in normal and pathological conditions. The present study examined FFA concentrations in CSF from 15 patients with traumatic brain injury (TBI) and compared the data with values obtained from 73 contemporary controls. Concentrations of specific FFAs from TBI patients, obtained within 48 h of the insult were significantly greater than those in the control group (arachidonic, docosahexaenoic and myristic, P<0.001; oleic, palmitic, P<0.01; linoleic, P<0.05). Higher concentrations of total polyunsaturated fatty acids (P<0.001) and of arachidonic, myristic and palmitic acids measured individually in CSF (P<0.01) obtained 1 week after the insult were associated with a worse outcome at the time of hospital discharge using the Glasgow Outcome Scale. This preliminary investigation suggests that CSF FFA concentrations may be useful as a predictive marker of outcome following TBI. PMID: 12946571 [PubMed - indexed for MEDLINE] 75. Prostaglandins Leukot Essent Fatty Acids. 2003 Oct;69(4):253-9. Potential role of dietary omega-3 essential fatty acids on some oxidant/antioxidant parameters in rats' corpus striatum. Sarsilmaz M, Songur A, Ozyurt H, Kuş I, Ozen OA, Ozyurt B, Söğüt S, Akyol O. Department of Anatomy, Faculty of Medicine, Firat University, Dekanlik Binasi, Elaziğ, Turkey. msarsilmaz@firat.edu.tr Omega-3 (omega-3) is an essential fatty acid (EFA) found in large amounts in fish oil. It contains eicosapentaenoic acid and docosahexaenoic acid (DHA). DHA is one of the building structures of membrane phospholipids of brain and necessary for continuity of neuronal functions. Evidences support the hypothesis that schizophrenia may be the result of increased reactive oxygen species mediated neuronal injury. Recent reports also suggest the protective effect of omega-3 EFA against neuropsychiatric disorders including schizophrenia. This study proposed to assess the changes in antioxidant enzyme and oxidant parameters in the corpus striatum (CS) of rats fed with omega-3 EFA diet (0.4g/kg/day) for 30 days. Eight control rats and nine rats fed with omega-3 were decapitated under ether anesthesia, and CS was removed immediately. Thiobarbituric acid-reactive substances (TBARS) and nitric oxide (NO) levels as well as total superoxide dismutase (t-SOD) and xanthine oxidase (XO) enzyme activities in the CS were measured. Rats treated with omega-3 EFA had significantly lower values of TBARS (P<0.001), NO (P<0.002) and XO (P<0.005) whereas higher values of t-SOD enzyme activity (P<0.002) than the control rats. These results indicate that omega-3 EFA rich fish oil diet reduces some oxidant parameters in CS. This may be revealed by means of reduced CS TBARS levels as an end product of lipid peroxidation of membranes in treated rats. Additionally, reduced XO activity and NO levels may support this notion. On the other hand, although the mechanism is not clear, omega-3 EFA may indirectly enhance the activity of antioxidant enzyme t-SOD. Taken together, this preliminary animal study provides strong support for a therapeutic effect of omega-3 EFA supplemented to classical neuroleptic regimen in the treatment of schizophrenic symptoms and tardive dyskinesia. PMID: 12907135 [PubMed - indexed for MEDLINE] 76. J Biol Chem. 2003 Apr 25;278(17):14677-87. Epub 2003 Feb 17. Novel docosatrienes and 17S-resolvins generated from docosahexaenoic acid in murine brain, human blood, and glial cells. Autacoids in anti-inflammation. Hong S, Gronert K, Devchand PR, Moussignac RL, Serhan CN. Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA. Docosahexaenoic acid (DHA, C22:6) is highly enriched in brain, synapses, and retina and is a major omega-3 fatty acid. Deficiencies in this essential fatty acid are reportedly associated with neuronal function, cancer, and inflammation. Here, using new lipidomic analyses employing high performance liquid chromatography coupled with a photodiode-array detector and a tandem mass spectrometer, a novel series of endogenous mediators was identified in blood, leukocytes, brain, and glial cells as 17S-hydroxy-containing docosanoids denoted as docosatrienes (the main bioactive member of the series was 10,17S-docosatriene) and 17S series resolvins. These novel mediators were biosynthesized via epoxide-containing intermediates and proved potent (pico- to nanomolar range) regulators of both leukocytes reducing infiltration in vivo and glial cells blocking their cytokine production. These results indicate that DHA is the precursor to potent protective mediators generated via enzymatic oxygenations to novel docosatrienes and 17S series resolvins that each regulate events of interest in inflammation and resolution. PMID: 12590139 [PubMed - indexed for MEDLINE] 77. Lipids. 2003 Apr;38(4):303-15. The potential role for arachidonic and docosahexaenoic acids in protection against some central nervous system injuries in preterm infants. Crawford MA, Golfetto I, Ghebremeskel K, Min Y, Moodley T, Poston L, Phylactos A, Cunnane S, Schmidt W. Institute of Brain Chemistry and Human Nutrition, The London Metropolitan University, North Campus, London N7 8DB, United Kingdom. michael@macrawf.demon.co.uk The risk of central nervous, visual, and auditory damage increases from 2/1000 live births in the normal birthweight to > 200/1000 as birthweight falls below 1500 g. Such babies are most likely to be born preterm. Advances in infant care have led to increasing numbers of very-low-birthweight, preterm infants surviving to school age with moderate to severe brain damage. Steroids are one of the current treatments, but they cause significant, long-term problems. The evidence reported here suggests an additional approach to protecting the very preterm infant by supporting neurovascular membrane integrity. The complications of preterm, very-low-birthweight babies include bronchopulmonary dysplasia, retinopathy of prematurity, intraventricular hemorrhage, periventricular leukomalacia, and necrotizing enterocolitis, all of which have a vascular component. Arachidonic acid (AA) and DHA are essential, structural, and functional constituents of cell membranes. They are especially required for the growth and function of the brain and vascular systems, which are the primary biofocus of human fetal growth. Molecular dynamics and experimental evidence suggest that DHA could be the ligand for the retinoid X receptor (RXR) in neural tissue. RXR activation is an obligatory step in signaling to the nucleus and in the regulation of gene expression. Very preterm babies are born with minimal fat stores and suboptimal circulating levels of these nutrients. Postnatally, they lose the biomagnification of the proportions of AA and DHA by the placenta for the fetus. No current nutritional management repairs these deficits. The placental biomagnification profile highlights AA rather than DHA. The resultant fetal FA profile closely resembles that of the vascular endothelium and not the brain. Without this nourishment, cell membrane abnormalities would be predicted. We present a scientific rationale for a common pathogenic process in the complications of prematurity. PMID: 12848275 [PubMed - indexed for MEDLINE] 78. Neuroscience. 2003;119(4):999-1012. Neuroprotective effect of developmental docosahexaenoic acid supplement against excitotoxic brain damage in infant rats. Högyes E, Nyakas C, Kiliaan A, Farkas T, Penke B, Luiten PG. Department of Animal Physiology, University of Groningen, Kerklaan 30, POB 14, 9750 AA, Haren, The Netherlands. Long-chain polyunsaturated fatty acid (LC-PUFA) composition of neural membranes is a key factor for brain development, in chemical communication of neurons and probably also their survival in response to injury. Viability of cholinergic neurons was tested during brain development following dietary supplementation of fish oil LC-PUFAs (docosahexaenoic acid [DHA], eicosapentaenoic acid, arachidonic acid) in the food of mother rats. Excitotoxic injury was introduced by N-methyl-D,L-aspartate (NMDA) injection into the cholinergic nucleus basalis magnocellularis of 14-day-old rats. The degree of loss of cholinergic cell bodies, and the extend of axonal and dendritic disintegration were measured following immunocytochemical staining of cell bodies and dendrites for choline acetyltransferase and p75 low-affinity neurotrophin receptor and by histochemical staining of acetylcholinesterase-positive fibres in the parietal neocortex. The impact of different feeding regimens on fatty acid composition of neural membrane phospholipids was also assayed at 12 days of age. Supplementation of LC-PUFAs resulted in a resistance against NMDA-induced excitotoxic degeneration of cholinergic neurones in the infant rats. More cholinergic cells survived, the dendritic involution of surviving neurons in the penumbra region decreased, and the degeneration of axons at the superficial layers of parietal neocortex also attenuated after supplementing LC-PUFAs. A marked increment in DHA content in all types of phospholipids was obtained in the forebrain neuronal membrane fraction of supplemented rats. It is concluded that fish oil LC-PUFAs, first of all DHA, is responsible for the neuroprotective action on developing cholinergic neurons against glutamate cytotoxicity. PMID: 12831859 [PubMed - indexed for MEDLINE] 79. J Neurochem. 2002 Nov;83(3):515-24. Systemic fatty acid responses to transient focal cerebral ischemia: influence of neuroprotectant therapy with human albumin. Rodriguez de Turco EB, Belayev L, Liu Y, Busto R, Parkins N, Bazan NG, Ginsberg MD. Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, School of Medicine, New Orleans, Louisiana, USA. Human albumin therapy is highly neuroprotective in focal cerebral ischemia. Because albumin is the main carrier of free fatty acids (FFA) in plasma, we investigated the content and composition of plasma FFA in jugular vein (JV), femoral artery (FA) and femoral vein (FV) of rats given intravenous human albumin (1.25 g/kg) or saline vehicle (5 mL/kg) 1 h after a 2 h middle cerebral artery occlusion (MCAo) or sham surgery. Arachidonic acid was the only FFA significantly increased by MCAo in all plasma samples prior to albumin administration, remaining at the same level regardless of subsequent treatments. Albumin treatment induced in both MCAo- and sham-groups a 1.7-fold increase in total plasma FFA (mainly 16:0, 18:1, 18:2n-6) during 90-min reperfusion. MCAo selectively stimulated the albumin-mediated mobilization of n-3 polyunsaturated fatty acids (PUFA), with an early increase in 22:5n-3 and 22:6n-3 in the FA prior to detectable changes in the JV. In the MCAo-albumin group, the lower level of FFA in JV as compared with FA and FV suggests an albumin-mediated systemic mobilization and supply of FFA to the brain, which may favor the replenishment of PUFA lost from cellular membranes during ischemia and/or to serve as an alternative source of energy, thus contributing to albumin neuroprotection. PMID: 12390513 [PubMed - indexed for MEDLINE] 80. J Exp Med. 2002 Oct 21;196(8):1025-37. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. Serhan CN, Hong S, Gronert K, Colgan SP, Devchand PR, Mirick G, Moussignac RL. Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA. cnserhan@zeus.bwh.harvard.edu Aspirin (ASA) is unique among current therapies because it acetylates cyclooxygenase (COX)-2 enabling the biosynthesis of R-containing precursors of endogenous antiinflammatory mediators. Here, we report that lipidomic analysis of exudates obtained in the resolution phase from mice treated with ASA and docosahexaenoic acid (DHA) (C22:6) produce a novel family of bioactive 17R-hydroxy-containing di- and tri-hydroxy-docosanoids termed resolvins. Murine brain treated with aspirin produced endogenous 17R-hydroxydocosahexaenoic acid as did human microglial cells. Human COX-2 converted DHA to 13-hydroxy-DHA that switched with ASA to 17R-HDHA that also proved a major route in hypoxic endothelial cells. Human neutrophils transformed COX-2-ASA-derived 17R-hydroxy-DHA into two sets of novel di- and trihydroxy products; one initiated via oxygenation at carbon 7 and the other at carbon 4. These compounds inhibited (IC(50) approximately 50 pM) microglial cell cytokine expression and in vivo dermal inflammation and peritonitis at ng doses, reducing 40-80% leukocytic exudates. These results indicate that exudates, vascular, leukocytes and neural cells treated with aspirin convert DHA to novel 17R-hydroxy series of docosanoids that are potent regulators. These biosynthetic pathways utilize omega-3 DHA and EPA during multicellular events in resolution to produce a family of protective compounds, i.e., resolvins, that enhance proresolution status. PMCID: PMC2194036 PMID: 12391014 [PubMed - indexed for MEDLINE] 81. Crit Care Clin. 1997 Jan;13(1):39-49. Nutritional management of the critically ill neurologic patient. Twyman D. Commercial Development Department, Nutrasweet Kelco Company, Deerfield, Illinois, USA. To summarize, the event of severe neurologic injury results in significant metabolic changes. These changes cause increased requirements for protein and nonprotein calories, micronutrients, and small bowel feedings or TPN. Early feeding has been shown to improve survival. Therefore, every effort should be made to provide aggressive nutritional support within the first 72 hours after injury. Specific guidelines are as follows: Provide full-strength, full-rate feedings within 72 hours. Provide enteral nutrients via nasojejunal or percutaneous endoscopic jejunostomy feeding tube if access is available; attempt gastric feedings if not. Provide TPN within 48 hours if enteral access is not available and begin enteral feeding as soon as possible. Provide 2 to 2.3 g protein/kg/d if renal function is normal. Provide 40% to 70% above basal needs as total calories, with 30% to 40% of calories as lipid to minimize hyperglycemia. Provide protein as small peptides to improve tolerance, absorption, utilization, and gut integrity. Provide a lipid source with 50% to 70% medium-chain triglycerides and an omega-6 to omega-3 ratio of 2:1 to 8:1 to minimize negative effects of omega-6 fatty acids and provide an easily absorbed and utilized source of lipid. PMID: 9012575 [PubMed - indexed for MEDLINE] 82. Neurology. 1993 Jun;43(6):1227-32. Effects of fish oil supplementation on acute ischemic brain injury in the rat. Lai ML, Hsu CY, Liu TH, He YY, Xu J, Navidi M, Sun G, Hogan EL. Department of Neurology, Medical University of South Carolina, Charleston. We studied the effects of long-term fish oil (FO) dietary supplementation on brain edema, polymorphonuclear neutrophil (PMN) infiltration, and infarct size in a rat stroke model. Rats were given regular rat chow with or without FO supplement (20% of total calories) for 7 weeks. Body weight did not differ between the two groups. In the FO group, an increase in eicosapentaenoic acid and a decrease in arachidonic acid content in hepatic phospholipids were significant in the phosphatidylcholine, phosphatidylethanolamine, and phosphatidylserine but not in the phosphoinositol fraction. Platelet activity reflected by serum thromboxane B2 levels was reduced in the FO group. Postischemic brain edema and PMN infiltration were not different between the two groups. The infarct volume was significantly greater in the FO group (controls: 96 +/- 7 mm3, n = 49; FO group: 124 +/- 6 mm3, n = 53; p = 0.0036). The greater ischemic brain injury in the FO-supplemented animals is probably related to the intraischemic hyperglycemia, which was worse in the FO group than in the control group (controls: 265 +/- 19 mg/dl, n = 14; FO group: 340 +/- 18 mg/dl, n = 16; p = 0.0079). PMID: 8170570 [PubMed - indexed for MEDLINE] 83. Thromb Res. 1992 Dec 1;68(4-5):383-91. The effect of dietary n-3 fatty acids on in vivo platelet aggregation in the cerebral microcirculation. Heizer ML, McKinney JS, Ellis EF. Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond 23298. Diets enriched in n-3 fish oil have been suggested to decrease coronary artery disease in part through their ability to decrease cyclooxygenase-dependent platelet aggregation. However little is known concerning the effect of n-3 fatty acids on in vivo platelet aggregation. The purpose of these experiments was to determine whether dietary n-3 fatty acids affect the rate at which platelet aggregation occurs in cerebral arterioles. Fish oil (200 mg eicosapentaenoic acid + 143 mg docosahexaenoic acid/kg), corn oil or water was given daily by gavage to mice (n = 30) for six weeks and then in vivo platelet aggregation was induced by the light plus dye method, which injuries the endothelium. Two additional groups of mice were acutely treated with saline or indomethacin (0.5 mg/kg, ip), with the latter serving as a positive control for therapeutic inhibition of platelet aggregation. Serum thromboxane B2 was analyzed by RIA. All fed groups gained weight equally. Serum thromboxane B2 was decreased by 40% in the fish oil group (p = 0.05 vs. corn oil, p = 0.07 vs. water). The mean (+/- SE) time to first aggregate in pial arterioles was 101 +/- 6, 91 +/- 6 and 101 +/- 9 seconds in the fish corn oil and water groups, respectively. Indomethacin significantly increased the time to first arteriolar aggregate by 35% (p < 0.002) and caused an 80% reduction in serum thromboxane. These studies show dietary fish oil produces a moderate reduction in serum TxB2 level and does not affect arteriolar platelet aggregation whereas indomethacin produces a drastic TxB2 reduction and significantly slows platelet aggregation.(ABSTRACT TRUNCATED AT 250 WORDS) PMID: 1290166 [PubMed - indexed for MEDLINE] 84. Proc Natl Acad Sci U S A. 1989 Apr;86(8):2903-7. Membrane docosahexaenoate is supplied to the developing brain and retina by the liver. Scott BL, Bazan NG. Louisiana State University School of Medicine, Louisiana State University Eye Center, New Orleans 70112. Docosahexaenoic acid [22:6 omega 3; 22:6(4, 7, 10, 13, 16, 19)] is concentrated in phospholipids of cellular membranes from brain and retina. Although linolenic acid [18:3 omega 3; 18:3(9, 12, 15)] is the major omega 3 fatty acid of mouse dams' milk, 22:6 is the prevalent omega 3 fatty acid in serum and tissues. Intraperitoneal injection of [1-14C]18:3 into 3-day-old mouse pups resulted in liver and serum lipid labeling that was initially high, followed by a rapid decline. In contrast, labeling of brain and retinal lipids were initially low and increased with time. Labeled 22:6 first appeared in liver 2 hr after injection and later in brain and retina. We suggest that 22:6 synthesized from 18:3 by the liver is secreted into the bloodstream in lipoproteins, taken up by brain and retina, and incorporated into cell membranes. We hypothesize that the 22:6 requirements of membranes (e.g., during synaptogenesis, photoreceptor membrane biogenesis, or repair after ischemic injury or neurodegenerative disorders) are met by a signal that is sent by the appropriate tissues to the liver to evoke the secretion of 22:6-containing lipoproteins. PMCID: PMC287028 PMID: 2523075 [PubMed - indexed for MEDLINE]