Skip to main content
The Journal of Nutrition logoLink to The Journal of Nutrition
. 2010 Apr;140(4):864–868. doi: 10.3945/jn.109.113233

DHA Deficiency and Prefrontal Cortex Neuropathology in Recurrent Affective Disorders1–3

Robert K McNamara 1,*
PMCID: PMC2838627  PMID: 20147466

Abstract

Increasing evidence suggests that docosahexaenoic acid [DHA, 22:6(n-3)], the principal (n-3) fatty acid in brain gray matter, has neurotrophic and neuroprotective properties. Preliminary clinical evidence also suggests that the perinatal accrual, and the subsequent dietary maintenance of, cortical DHA is positively associated with cortical gray matter volumes. The pathophysiology of recurrent affective disorders, including unipolar and bipolar depression, is associated with (n-3) fatty acid deficiency, DHA deficits, impaired astrocyte mediated vascular coupling, neuronal shrinkage, and reductions in gray matter volume in the prefrontal cortex (PFC). Preclinical studies have also observed neuronal shrinkage and indices of astrocyte pathology in the DHA-deficient rat brain. Together, this body of evidence supports the proposition that DHA deficiency increases vulnerability to neuronal atrophy in the PFC of patients with affective disorders. Because projections from the PFC modulate multiple limbic structures involved in affective regulation, this represents one plausible mechanism by which (n-3) fatty acid deficiency may increase vulnerability to recurrent affective disorders.

Introduction

The WHO identified major depressive disorder (MDD)5 as the 4th ranked cause of disability and premature death globally (1) and excess mortality in MDD is primarily attributable to suicide and cardiovascular-related disorders (2). It has been projected that by 2020, MDD and ischemic heart disease will be the 2 most important causes of disability worldwide (1). In the US, lifetime prevalence rates are currently estimated at 16% for MDD and 1.1% for bipolar disorders (3). Subtotal heritability estimates and monozygotic twin discordance rates indicate that nongenetic factors confer equal if not greater vulnerability for recurrent affective disorders, including MDD and bipolar disorder (4). Accordingly, efforts need to be made to identify environmental risk factors that confer vulnerability, particularly in view of their amenability to prevention.

A growing body of evidence from cross-national and cross-sectional epidemiological surveys and prospective longitudinal studies suggest that greater habitual dietary (n-3) fatty acid intake is associated with reduced risk for both MDD and bipolar disorder (57). This is further supported by independent meta-analyses of placebo-controlled trials finding that increasing dietary intake of (n-3) fatty acids, including docosahexaenoic acid [DHA, 22:6(n-3)] and/or eicosapentaenoic acid [EPA, 20:5(n-3)], is associated with a significant advantage over placebo for reducing depression symptom severity in adult patients with affective disorders (8,9). Subsequent trials have also found that increasing dietary (n-3) fatty acid intake is associated with significant reductions in depression and manic symptom severity in pediatric and adolescent patients (1012). Dietary (n-3) fatty acid intake is reflected in erythrocyte membrane (n-3) fatty acid composition, and case-control studies have repeatedly observed significant erythrocyte (n-3) fatty acid deficits in patients with MDD and bipolar disorder (1316). Although these data suggest that (n-3) fatty acid deficiency represents a reversible environmental risk factor for affective disorders, it remains unclear how (n-3) fatty acid deficiency increases vulnerability to affective dysregulation.

DHA [22:6(n-3)] is the principal (n-3) fatty acid in cortical gray matter, accounting for ∼15% of total fatty acid composition in the adult human prefrontal cortex (PFC) (17,18). Precursors of DHA, including EPA and docosapentaenoic acid [22:5(n-3)], comprise <1% of total brain fatty acid composition. Recent postmortem studies have found significant DHA deficits in PFC gray matter of patients with affective disorders (19,20). Because preclinical studies have found that brain DHA and/or its bioactive metabolites are neuroprotective (21), DHA deficits in PFC gray matter may increase vulnerability to the neuropathology also observed in the PFC of patients with affective disorders (22). In this paper, evidence linking DHA deficiency and neuropathology in the PFC is summarized and evaluated as a potential mechanism by which (n-3) fatty acid deficiency may contribute vulnerability to affective dysregulation.

DHA: neurotrophic and neuroprotective properties

During perinatal development of the human brain, cortical concentrations of DHA increase sharply in association with active periods of neurogenesis, neuroblast migration, differentiation, and synaptogenesis (17,23). Preclinical studies have demonstrated that DHA positively regulates cortical neurogenesis (24), neuroblast migration (25), neuronal differentiation/arborization (26), neurotrophic factor expression (27), and nerve growth factor-induced neurite outgrowth and synaptogenesis (28). These developmental studies suggest that DHA has important neurotrophic properties required for normal perinatal cortical maturation. Indeed, dietary (n-3) fatty acid insufficiency during perinatal rat brain development is associated with enduring alterations in dopamine and serotonin neurotransmission (29), cognitive deficits, and elevated behavioral indices of anxiety, aggression, and depression (30) in young adulthood.

Dietary (n-3) fatty acid supplementation resulting in elevations in brain DHA content has been found to be neuroprotective against a variety of insults associated with the generation of reactive oxygen species and oxidative stress, including focal and global ischemia (31), glutamate excitotoxicity (32,33), and traumatic head injury (34). Furthermore, (n-3) fatty acid supplementation prevents or significantly attenuates age-related neuropathological features in a transgenic mouse model (APPswe) of Alzheimer's disease, including amyloid-β deposition, caspase activity, presenilin expression, dendritic pathology, plaque burden, and neuronal apotosis (35,36). Emerging data also suggest that increasing brain DHA content during perinatal rodent development increases neuronal resilience to oxidative stress and indices of lipid peroxidation (37,38), and perinatal (n-3) fatty acid insufficiency resulting in significant cortical DHA deficits is associated with region-specific reductions in neuron size (39). The mechanisms mediating the neuroprotective effects of DHA and its lipoxygenase metabolite, Neuroprotectin D1, may involve several complementary pathways, including reductions in proinflammatory signaling [prostaglandin E2 synthesis], upregulating antiapoptotic genes (Bcl-2, Bcl-xl, Bfl-1), and decreasing proapoptotic genes (Bax, Bad) (40). Importantly, elevated markers of excitotoxicity and inflammation are observed in the postmortem PFC of bipolar patients (41). Moreover, mood stabilizers including lithium also converge on these signaling pathways and, like DHA, have neurotrophic (42) and neuroprotective (43) properties. Mood stabilizer medications as a class also have antiinflammatory properties mediated in part by decreasing arachidonic acid [20:4(n-6)] generated proinflammatory signaling cascades (44).

Preliminary clinical data also suggest that (n-3) fatty acids promote neuronal resilience in humans. First, human and primate infants born preterm exhibit significant deficits in cortical DHA composition (4548) and neuroimaging studies have found that children and adolescents born preterm exhibit significant reductions in regional cortical gray matter volumes (49,50). Second, a voxel-based morphometry study found that habitual dietary (n-3) fatty acid intake was positively correlated with anterior cingulate and amygdala gray matter volumes among 55 healthy adult participants residing in the US (51). Third, a preliminary double-blind, placebo-controlled neuroimaging (proton MRI) trial found that chronic (12 wk) EPA [20:5(n-3)] treatment significantly increased cortical concentrations of N-acetyl aspartate, a putative measure of neuronal integrity, in the anterior cingulate (single voxel) of adult bipolar patients (52). Fourth, the orbitofrontal cortex (OFC) subregion of the PFC exhibits significant reductions in gray matter volume with increasing age (53) and DHA composition exhibits a significant age-related decline in the postmortem OFC (18). These clinical findings suggest that the perinatal accrual of cortical DHA, and the subsequent maintenance of cortical DHA through the diet, is positively associated with PFC gray matter volume over the human lifespan.

Neuropathology in affective disorders

Evidence from neuroimaging and postmortem histology studies indicate that adult patients with affective disorders have significant neuropathology in the PFC. Among the different PFC subregions, the OFC (Brodmann Areas 10,11,47) has emerged as a region that is particularly vulnerable to gray matter volume deficits in young adult and geriatric patients with affective disorders (5460). Postmortem histological studies suggest that deficits in PFC gray matter volume in young adult and elderly patients with affective disorders are attributable in part to pyramidal neuron atrophy (i.e. reductions in neuronal size and density) (6164). In addition to deficits in gray matter density, bipolar patients also have significant frontal white matter tract pathology (61). Importantly, glutamatergic pyramidal neurons within the human OFC have reciprocal connections with limbic structures, including the amygdala, hippocampus, hypothalamus, and striatum (65) (Fig. 1), and evidence from lesion and imaging studies suggest that the OFC modulates hedonic and emotional processes that characterize affective disorders, including euphoria, depression, irritability, impulsivity, recklessness, and socially inappropriate behavior (66). The pathophysiological relevance of OFC gray matter volume deficits in affective disorders is further supported by associations with depression symptom severity (67). For example, chronic (4 wk) treatment with lithium was associated with a significant increase in PFC gray matter volume only in bipolar patients exhibiting a significant decrease in depression severity (68).

FIGURE 1 .

FIGURE 1 

Diagram illustrating the connections between the prefrontal cortex (Brodmann Areas 10, 11, 47) and subcortical structures implicated in affective regulation. Potential consequences of abnormal functional connectivity between the prefrontal cortex and these regions are presented, including dysregulation in hypothalamic-pituitary-adrenal axis (cortisol) and mesolimbic dopamine activity. A dysregulation in these processes are major features associated with the pathophysiology of mood symptoms in recurrent affective disorders.

Although the etiological factors contributing to PFC neuropathology in affective disorders are not known, the absence of reactive gliosis is consistent with a developmental etiology rather than neurodegenerative processes (68). Indeed, longitudinal and cross-sectional structural imaging studies have found that affective disorders are associated with excessive gray matter loss in the OFC initiating during adolescence, when the first onset of affective disorders typically occurs (70,71). Specifically, OFC gray matter deficits are commonly observed in young adult and elderly patients with affective disorders (5460) but not in first-episode (72) or adolescent patients with affective disorders (73,74). A recent 2-y longitudinal study found that OFC gray matter volume reductions during adolescence were accelerated in bipolar patients compared with healthy subjects (75). In this study, the relative OFC gray matter volume loss in typical adolescents (0.33%/y) was increased 5-fold in bipolar patients (1.8%/y). It is relevant, therefore, that the normal age-related increase in PFC DHA composition occurring between adolescence and young adulthood in healthy subjects (17) was not observed in the OFC of suicide victims with or without a history of affective disorders (76,77). Together, these data suggest that deficits in OFC DHA accrual during adolescence may contribute to accelerated and excessive gray matter loss in affective disorders.

Glial pathology in affective disorders

In addition to neuronal atrophy, several studies have observed reductions in glial size and density in the postmortem PFC of patients with MDD or bipolar disorder (69). Subsequent studies have found reductions in the expression of astrocyte-specific marker glial fibrillary acidic protein in the PFC of young, but not older, patients with MDD (78,79). Additionally, an immunohistochemical study found a significant paucity of astrocytes adjacent to blood vessels in the PFC of MDD patients (80). These findings are important, because astrocytes play a critical role in the vascular uptake and metabolism of glucose (81,82). Furthermore, the expression of the astrocyte-specific glucose transporter (45-kDa GLUT1) (83) and glucose uptake (84) are significantly reduced in the PFC of DHA-deficient rats. Moreover, we found a significant reduction in myo-inositol, a metabolite of glucose concentrated in astrocytes, in the PFC of DHA-deficient rats by magnetic resonance imaging (85), and significantly lower myo-inositol concentrations are also observed in the PFC of patients with unipolar or bipolar depression (86,87). In contrast, increasing dietary (n-3) fatty acid intake was found to significantly improve age-related deficits in cerebrovascular coupling in nonhuman primates (88) and we found that 8-wk DHA supplementation significantly increased PFC activation during sustained attention in healthy pediatric human participants (89). Lastly, plasma DHA levels were found to be associated with regional brain glucose metabolism in patients with MDD (90). Together, these data suggest that DHA deficiency impairs astrocyte-mediated neuron-vascular coupling, which may in turn contribute to neuronal atrophy in the PFC of patients with affective disorders.

Summary and conclusions

There is now considerable evidence that DHA and/or its bioactive metabolites have neurotrophic and neuroprotective properties and that DHA increases neuronal resilience to oxidative stress during perinatal development and in adulthood. Preclinical findings also indicate that dietary-induced DHA deficits in rat brain are sufficient to produce neuropathological features, including neuronal shrinkage and astrocyte pathology, also observed in the PFC of patients with affective disorders. Clinical evidence also suggests that cortical DHA accrual is positively associated with gray matter volumes and that affective disorders are associated with deficits in PFC DHA composition, gray matter volume, and neuronal pathology. This body of evidence supports the proposal that cortical DHA deficits in patients with affective disorders reduce neuronal resilience in the PFC to oxidative stress. Because projections from the PFC modulate multiple limbic structures involved in affective regulation, including the amygdala, this represents one plausible mechanism by which (n-3) fatty acid deficiency may increase vulnerability to affective disorders. However, direct support for this pathoetiological mechanism will require evidence that (n-3) fatty acid supplementation during the prodromal phase of illness prevents the emergence of PFC gray matter volume deficits and affective dysregulation in asymptomatic adolescents with a familial risk for developing affective disorders.

Acknowledgments

R.K.M. wrote the paper and had primary responsibility for the final content.

1

Published as a supplement to The Journal of Nutrition. Presented as part of the symposium entitled “DHA and Neurodegenerative Disease: Models of Investigation” given at the Experimental Biology 2009 meeting, April 19, 2009, in New Orleans, LA. This symposium was sponsored the ASN and supported by an unrestricted educational grant from Martek Bioscience. The symposium was chaired by Jay Whelan and Robert K. McNamara. Guest Editor for this symposium publication was Cathy Levenson. Guest Editor disclosure: no conflicts to disclose.

2

Supported in part by National Institute of Mental Health grant MH083924.

3

Author disclosure: R. K. McNamara received funding from Martek Biosciences Inc., the Inflammation Research Foundation, Janssen Ortho-McNeil Pharmaceuticals, and NIH/National Institute of Mental Health.

5

Abbreviations used: DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; MDD, major depressive disorder; OFC, orbitofrontal cortex; PFC, prefrontal cortex.

References

  • 1.WHO. Cross-national comparisons of the prevalences and correlates of mental disorders. WHO International Consortium in Psychiatric Epidemiology. Bull World Health Organ. 2000;78:413–26. [PMC free article] [PubMed] [Google Scholar]
  • 2.Angst F, Stassen HH, Clayton PJ, Angst J. Mortality of patients with mood disorders: follow-up over 34–38 years. J Affect Disord. 2002;68:167–81. [DOI] [PubMed] [Google Scholar]
  • 3.Kessler RC, Merikangas KR, Wang PS. Prevalence, comorbidity, and service utilization for mood disorders in the United States at the beginning of the twenty-first century. Annu Rev Clin Psychol. 2007;3:137–58. [DOI] [PubMed] [Google Scholar]
  • 4.Merikangas KR, Chakravarti A, Moldin SO, Araj H, Blangero JC, Burmeister M, Crabbe J Jr, Depaulo JR Jr, Foulks E, et al. Future of genetics of mood disorders research. Biol Psychiatry. 2002;52:457–77. [DOI] [PubMed] [Google Scholar]
  • 5.McNamara RK. Evidence-based evaluation of omega-3 fatty acid deficiency as a risk factor for recurrent neuropsychiatric illness: current status and future directions. In: Heikkinen EP, editor. Fish oils and health. New York: Nova Science Publishers; 2008. p. 7–67.
  • 6.Hibbeln JR. Depression, suicide and deficiencies of omega-3 essential fatty acids in modern diets. World Rev Nutr Diet. 2009;99:17–30. [DOI] [PubMed] [Google Scholar]
  • 7.Parker G, Gibson NA, Brotchie H, Heruc G, Rees AM, Hadzi-Pavlovic D. Omega-3 fatty acids and mood disorders. Am J Psychiatry. 2006;163:969–78. [DOI] [PubMed] [Google Scholar]
  • 8.Freeman MP, Hibbeln JR, Wisner KL, Davis JM, Mischoulon D, Peet M, Keck PE Jr, Marangell LB, Richardson AJ, et al. Omega-3 fatty acids: evidence basis for treatment and future research in psychiatry. J Clin Psychiatry. 2006;67:1954–67. [DOI] [PubMed] [Google Scholar]
  • 9.Lin PY, Su KP. A meta-analytic review of double-blind, placebo-controlled trials of antidepressant efficacy of omega-3 fatty acids. J Clin Psychiatry. 2007;68:1056–61. [DOI] [PubMed] [Google Scholar]
  • 10.Clayton EH, Hanstock TL, Hirneth SJ, Kable CJ, Garg ML, Hazell PL. Reduced mania and depression in juvenile bipolar disorder associated with long-chain omega-3 polyunsaturated fatty acid supplementation. Eur J Clin Nutr. 2009;63:1037–40. [DOI] [PubMed] [Google Scholar]
  • 11.Nemets H, Nemets B, Apter A, Bracha Z, Belmaker RH. Omega-3 treatment of childhood depression: a controlled, double-blind pilot study. Am J Psychiatry. 2006;163:1098–100. [DOI] [PubMed] [Google Scholar]
  • 12.Wozniak J, Biederman J, Mick E, Waxmonsky J, Hantsoo L, Best C, Cluette-Brown JE, Laposata M. Omega-3 fatty acid monotherapy for pediatric bipolar disorder: a prospective open-label trial. Eur Neuropsychopharmacol. 2007;17:440–7. [DOI] [PubMed] [Google Scholar]
  • 13.Chiu CC, Huang SY, Su KP, Lu ML, Huang MC, Chen CC, Shen WW. Polyunsaturated fatty acid deficit in patients with bipolar mania. Eur Neuropsychopharmacol. 2003;13:99–103. [DOI] [PubMed] [Google Scholar]
  • 14.Edwards R, Peet M, Shay J, Horrobin D. Omega-3 polyunsaturated fatty acid levels in the diet and in red blood cell membranes of depressed patients. J Affect Disord. 1998;48:149–55. [DOI] [PubMed] [Google Scholar]
  • 15.McNamara RK, Jandacek R, Rider T, Tso P, Dwivedi Y, Pandey GN. Selective erythrocyte omega-3 fatty acid deficits in adult patients with bipolar disorder: comparison with major depressive disorder. Meeting of the Society of Biological Psychiatry. 2009;64:140S. [Google Scholar]
  • 16.Peet M, Murphy B, Shay J, Horrobin D. Depletion of omega-3 fatty acid levels in red blood cell membranes of depressive patients. Biol Psychiatry. 1998;43:315–9. [DOI] [PubMed] [Google Scholar]
  • 17.Carver JD, Benford VJ, Han B, Cantor AB. The relationship between age and the fatty acid composition of cerebral cortex and erythrocytes in human subjects. Brain Res Bull. 2001;56:79–85. [DOI] [PubMed] [Google Scholar]
  • 18.McNamara RK, Liu Y, Jandacek R, Rider T, Tso P. The aging human orbitofrontal cortex: decreasing polyunsaturated fatty acid composition and associated increases in lipogenic gene expression and stearoyl-CoA desaturase activity. Prostaglandins Leukot Essent Fatty Acids. 2008;78:293–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.McNamara RK, Hahn CG, Jandacek R, Rider T, Tso P, Stanford KE, Richtand NM. Selective deficits in the omega-3 fatty acid docosahexaenoic acid in the postmortem orbitofrontal cortex of patients with major depressive disorder. Biol Psychiatry. 2007;62:17–24. [DOI] [PubMed] [Google Scholar]
  • 20.McNamara RK, Jandacek R, Rider T, Tso P, Stanford KE, Hahn CG, Richtand NM. Deficits in docosahexaenoic acid and associated elevations in the metabolism of arachidonic acid and saturated fatty acids in the postmortem orbitofrontal cortex of patients with bipolar disorder. Psychiatry Res. 2008;160:285–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bazan NG. Omega-3 fatty acids, pro-inflammatory signaling and neuroprotection. Curr Opin Clin Nutr Metab Care. 2007;10:136–41. [DOI] [PubMed] [Google Scholar]
  • 22.Rajkowska G. Cell pathology in mood disorders. Semin Clin Neuropsychiatry. 2002;7:281–92. [DOI] [PubMed] [Google Scholar]
  • 23.Clandinin MT, Chappell JE, Leong S, Heim T, Swyer PR, Chance GW. Extrauterine fatty acid accretion in infant brain: implications for fatty acid requirements. Early Hum Dev. 1980;4:131–8. [DOI] [PubMed] [Google Scholar]
  • 24.Coti Bertrand P, O'Kusky JR, Innis SM. Maternal dietary (n-3) fatty acid deficiency alters neurogenesis in the embryonic rat brain. J Nutr. 2006;136:1570–5. [DOI] [PubMed] [Google Scholar]
  • 25.Yavin E, Himovichi E, Eilam R. Delayed cell migration in the developing rat brain following maternal omega 3 alpha linolenic acid dietary deficiency. Neuroscience. 2009;162:1011–22. [DOI] [PubMed] [Google Scholar]
  • 26.Calderon F, Kim HY. Docosahexaenoic acid promotes neurite growth in hippocampal neurons. J Neurochem. 2004;90:979–88. [DOI] [PubMed] [Google Scholar]
  • 27.Ikemoto A, Nitta A, Furukawa S, Ohishi M, Nakamura A, Fujii Y, Okuyama H. Dietary n-3 fatty acid deficiency decreases nerve growth factor content in rat hippocampus. Neurosci Lett. 2000;285:99–102. [DOI] [PubMed] [Google Scholar]
  • 28.Martin RE, Bazan NG. Changing fatty acid content of growth cone lipids prior to synaptogenesis. J Neurochem. 1992;59:318–25. [DOI] [PubMed] [Google Scholar]
  • 29.Chalon S. Omega-3 fatty acids and monoamine neurotransmission. Prostaglandins Leukot Essent Fatty Acids. 2006;75:259–69. [DOI] [PubMed] [Google Scholar]
  • 30.Fedorova I, Salem N Jr. Omega-3 fatty acids and rodent behavior. Prostaglandins Leukot Essent Fatty Acids. 2006;75:271–89. [DOI] [PubMed] [Google Scholar]
  • 31.Belayev L, Khoutorova L, Atkins KD, Bazan NG. Robust docosahexaenoic acid-mediated neuroprotection in a rat model of transient, focal cerebral ischemia. Stroke. 2009;40:3121–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Blondeau N, Widmann C, Lazdunski M, Heurteaux C. Polyunsaturated fatty acids induce ischemic and epileptic tolerance. Neuroscience. 2002;109:231–41. [DOI] [PubMed] [Google Scholar]
  • 33.Ozyurt B, Sarsilmaz M, Akpolat N, Ozyurt H, Akyol O, Herken H, Kus I. The protective effects of omega-3 fatty acids against MK-801-induced neurotoxicity in prefrontal cortex of rat. Neurochem Int. 2007;50:196–202. [DOI] [PubMed] [Google Scholar]
  • 34.Wu A, Ying Z, Gomez-Pinilla F. Dietary omega-3 fatty acids normalize BDNF levels, reduce oxidative damage, and counteract learning disability after traumatic brain injury in rats. J Neurotrauma. 2004;21:1457–67. [DOI] [PubMed] [Google Scholar]
  • 35.Cole GM, Frautschy SA. Docosahexaenoic acid protects from amyloid and dendritic pathology in an Alzheimer's disease mouse model. Nutr Health. 2006;18:249–59. [DOI] [PubMed] [Google Scholar]
  • 36.Green KN, Martinez-Coria H, Khashwji H, Hall EB, Yurko-Mauro KA, Ellis L, LaFerla FM. Dietary docosahexaenoic acid and docosapentaenoic acid ameliorate amyloid-beta and tau pathology via a mechanism involving presenilin 1 levels. J Neurosci. 2007;27:4385–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Green P, Glozman S, Weiner L, Yavin E. Enhanced free radical scavenging and decreased lipid peroxidation in the rat fetal brain after treatment with ethyl docosahexaenoate. Biochim Biophys Acta. 2001;1532:203–12. [DOI] [PubMed] [Google Scholar]
  • 38.Högyes E, Nyakas C, Kiliaan A, Farkas T, Penke B, Luiten PG. Neuroprotective effect of developmental docosahexaenoic acid supplement against excitotoxic brain damage in infant rats. Neuroscience. 2003;119:999–1012. [DOI] [PubMed] [Google Scholar]
  • 39.Ahmad A, Moriguchi T, Salem N. Decrease in neuron size in docosahexaenoic acid-deficient brain. Pediatr Neurol. 2002;26:210–8. [DOI] [PubMed] [Google Scholar]
  • 40.Bazan NG. Cellular and molecular events mediated by docosahexaenoic acid-derived neuroprotectin D1 signaling in photoreceptor cell survival and brain protection. Prostaglandins Leukot Essent Fatty Acids. 2009;81:205–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rao JS, Harry GJ, Rapoport SI, Kim HW. Increased excitotoxicity and neuroinflammatory markers in postmortem frontal cortex from bipolar disorder patients. Mol Psychiatry. Epub 2009. Jun 2. [DOI] [PMC free article] [PubMed]
  • 42.Shaltiel G, Chen G, Manji HK. Neurotrophic signaling cascades in the pathophysiology and treatment of bipolar disorder. Curr Opin Pharmacol. 2007;7:22–6. [DOI] [PubMed] [Google Scholar]
  • 43.Bachmann RF, Schloesser RJ, Gould TD, Manji HK. Mood stabilizers target cellular plasticity and resilience cascades: implications for the development of novel therapeutics. Mol Neurobiol. 2005;32:173–202. [DOI] [PubMed] [Google Scholar]
  • 44.Rao JS, Lee HJ, Rapoport SI, Bazinet RP. Mode of action of mood stabilizers: is the arachidonic acid cascade a common target? Mol Psychiatry. 2008;13:585–96. [DOI] [PubMed] [Google Scholar]
  • 45.Clandinin MT, Chappell JE, Leong S, Heim T, Swyer PR, Chance GW. Intrauterine fatty acid accretion rates in human brain: implications for fatty acid requirements. Early Hum Dev. 1980;4:121–9. [DOI] [PubMed] [Google Scholar]
  • 46.Farquharson J, Cockburn F, Patrick WA, Jamieson EC, Logan RW. Infant cerebral cortex phospholipid fatty-acid composition and diet. Lancet. 1992;340:810–3. [DOI] [PubMed] [Google Scholar]
  • 47.Martinez M. Tissue levels of polyunsaturated fatty acids during early human development. J Pediatr. 1992;120:S129–38. [DOI] [PubMed] [Google Scholar]
  • 48.Sarkadi-Nagy E, Wijendran V, Diau GY, Chao AC, Hsieh AT, Turpeinen A, Nathanielsz PW, Brenna JT. The influence of prematurity and long chain polyunsaturate supplementation in 4-week adjusted age baboon neonate brain and related tissues. Pediatr Res. 2003;54:244–52. [DOI] [PubMed] [Google Scholar]
  • 49.Nosarti C, Al-Asady MH, Frangou S, Stewart AL, Rifkin L, Murray RM. Adolescents who were born very preterm have decreased brain volumes. Brain. 2002;125:1616–23. [DOI] [PubMed] [Google Scholar]
  • 50.Peterson BS, Vohr B, Staib LH, Cannistraci CJ, Dolberg A, Schneider KC, Katz KH, Westerveld M, Sparrow S, et al. Regional brain volume abnormalities and long-term cognitive outcome in preterm infants. JAMA. 2000;284:1939–47. [DOI] [PubMed] [Google Scholar]
  • 51.Conklin SM, Gianaros PJ, Brown SM, Yao JK, Hariri AR, Manuck SB, Muldoon MF. Long-chain omega-3 fatty acid intake is associated positively with corticolimbic gray matter volume in healthy adults. Neurosci Lett. 2007;421:209–12. [DOI] [PubMed] [Google Scholar]
  • 52.Frangou S, Lewis M, Wollard J, Simmons A. Preliminary in vivo evidence of increased N-acetyl-aspartate following eicosapentanoic acid treatment in patients with bipolar disorder. J Psychopharmacol. 2007;21:435–9. [DOI] [PubMed] [Google Scholar]
  • 53.Raz N, Gunning FM, Head D, Dupuis JH, McQuain J, Briggs SD, Loken WJ, Thornton AE, Acker JD. Selective aging of the human cerebral cortex observed in vivo: differential vulnerability of the prefrontal gray matter. Cereb Cortex. 1997;7:268–82. [DOI] [PubMed] [Google Scholar]
  • 54.Ballmaier M, Toga AW, Blanton RE, Sowell ER, Lavretsky H, Peterson J, Pham D, Kumar A. Anterior cingulate, gyrus rectus, and orbitofrontal abnormalities in elderly depressed patients: an MRI-based parcellation of the prefrontal cortex. Am J Psychiatry. 2004;161:99–108. [DOI] [PubMed] [Google Scholar]
  • 55.Bremner JD, Vythilingam M, Vermetten E, Nazeer A, Adil J, Khan S, Staib LH, Charney DS. Reduced volume of orbitofrontal cortex in major depression. Biol Psychiatry. 2002;51:273–9. [DOI] [PubMed] [Google Scholar]
  • 56.Lacerda AL, Keshavan MS, Hardan AY, Yorbik O, Brambilla P, Sassi RB, Nicoletti M, Mallinger AG, Frank E, et al. Anatomic evaluation of the orbitofrontal cortex in major depressive disorder. Biol Psychiatry. 2004;55:353–8. [DOI] [PubMed] [Google Scholar]
  • 57.Lai T, Payne ME, Byrum CE, Steffens DC, Krishnan KR. Reduction of orbital frontal cortex volume in geriatric depression. Biol Psychiatry. 2000;48:971–5. [DOI] [PubMed] [Google Scholar]
  • 58.Lee SH, Payne ME, Steffens DC, McQuoid DR, Lai TJ, Provenzale JM, Krishnan KR. Subcortical lesion severity and orbitofrontal cortex volume in geriatric depression. Biol Psychiatry. 2003;54:529–33. [DOI] [PubMed] [Google Scholar]
  • 59.Stanfield AC, Moorhead TW, Job DE, McKirdy J, Sussmann JE, Hall J, Giles S, Johnstone EC, Lawrie SM, et al. Structural abnormalities of ventrolateral and orbitofrontal cortex in patients with familial bipolar disorder. Bipolar Disord. 2009;11:135–44. [DOI] [PubMed] [Google Scholar]
  • 60.Beyer JL, Kuchibhatla M, Payne ME, Macfall J, Cassidy F, Krishnan KR. Gray and white matter brain volumes in older adults with bipolar disorder. Int J Geriatr Psychiatry. 2009;24:1445–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Rajkowska G, Miguel-Hidalgo JJ, Dubey P, Stockmeier CA, Krishnan KR. Prominent reduction in pyramidal neurons density in the orbitofrontal cortex of elderly depressed patients. Biol Psychiatry. 2005;58:297–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Van Otterloo E, O'Dwyer G, Stockmeier CA, Steffens DC, Krishnan RR, Rajkowska G. Reductions in neuronal density in elderly depressed are region specific. Int J Geriatr Psychiatry. 2009;24:856–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Cotter D, Hudson L, Landau S. Evidence for orbitofrontal pathology in bipolar disorder and major depression, but not in schizophrenia. Bipolar Disord. 2005;7:358–69. [DOI] [PubMed] [Google Scholar]
  • 64.Rajkowska G, Miguel-Hidalgo JJ, Wei J, Dilley G, Pittman SD, Meltzer HY, Overholser JC, Roth BL, Stockmeier CA. Morphometric evidence for neuronal and glial prefrontal cell pathology in major depression. Biol Psychiatry. 1999;45:1085–98. [DOI] [PubMed] [Google Scholar]
  • 65.Kafantaris V, Kingsley P, Ardekani B, Saito E, Lencz T, Lim K, Szeszko P. Lower orbital frontal white matter integrity in adolescents with bipolar I disorder. J Am Acad Child Adolesc Psychiatry. 2009;48:79–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kringelbach ML, Rolls ET. The functional neuroanatomy of the human orbitofrontal cortex: evidence from neuroimaging and neuropsychology. Prog Neurobiol. 2004;72:341–72. [DOI] [PubMed] [Google Scholar]
  • 67.Kringelbach ML. The human orbitofrontal cortex: linking reward to hedonic experience. Nat Rev Neurosci. 2005;6:691–702. [DOI] [PubMed] [Google Scholar]
  • 68.Nery FG, Chen HH, Hatch JP, Nicoletti MA, Brambilla P, Sassi RB, Mallinger AG, Keshavan MS, Soares JC. Orbitofrontal cortex gray matter volumes in bipolar disorder patients: a region-of-interest MRI study. Bipolar Disord. 2009;11:145–53. [DOI] [PubMed] [Google Scholar]
  • 69.Moore GJ, Cortese BM, Glitz DA, Zajac-Benitez C, Quiroz JA, Uhde TW, Drevets WC, Manji HK. A longitudinal study of the effects of lithium treatment on prefrontal and subgenual prefrontal gray matter volume in treatment-responsive bipolar disorder patients. J Clin Psychiatry. 2009;70:699–705. [DOI] [PubMed] [Google Scholar]
  • 70.Cotter DR, Pariante CM, Everall IP. Glial cell abnormalities in major psychiatric disorders: the evidence and implications. Brain Res Bull. 2001;55:585–95. [DOI] [PubMed] [Google Scholar]
  • 71.Perlis RH, Miyahara S, Marangell LB, Wisniewski SR, Ostacher M, DelBello MP, Bowden CL, Sachs GS, Nierenberg AA, STEP-BD Investigators. Long-term implications of early onset in bipolar disorder: data from the first 1000 participants in the systematic treatment enhancement program for bipolar disorder (STEP-BD). Biol Psychiatry. 2004;55:875–81. [DOI] [PubMed] [Google Scholar]
  • 72.Burke KC, Burke JD Jr, Rae DS, Regier DA. Comparing age at onset of major depression and other psychiatric disorders by birth cohorts in five US community populations. Arch Gen Psychiatry. 1991;48:789–95. [DOI] [PubMed] [Google Scholar]
  • 73.Adler CM, DelBello MP, Jarvis K, Levine A, Adams J, Strakowski SM. Voxel-based study of structural changes in first-episode patients with bipolar disorder. Biol Psychiatry. 2007;61:776–81. [DOI] [PubMed] [Google Scholar]
  • 74.Blumberg HP, Krystal JH, Bansal R, Martin A, Dziura J, Durkin K, Martin L, Gerard E, Charney DS, et al. Age, rapid-cycling, and pharmacotherapy effects on ventral prefrontal cortex in bipolar disorder: a cross-sectional study. Biol Psychiatry. 2006;59:611–8. [DOI] [PubMed] [Google Scholar]
  • 75.Chen HH, Rosenberg DR, MacMaster FP, Easter PC, Caetano SC, Nicoletti M, Hatch JP, Nery FG, Soares JC. Orbitofrontal cortex volumes in medication naïve children with major depressive disorder: a magnetic resonance imaging study. J Child Adolesc Psychopharmacol. 2008;18:551–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kalmar JH, Wang F, Spencer L, Edmiston E, Lacadie CM, Martin A, Constable RT, Duncan JS, Staib LH, et al. Preliminary evidence for progressive prefrontal abnormalities in adolescents and young adults with bipolar disorder. J Int Neuropsychol Soc. 2009;15:476–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.McNamara RK, Jandacek R, Rider T, Tso P, Dwivedi Y, Roberts RC, Conley RR, Pandey GN. Fatty acid composition of the postmortem prefrontal cortex of male and female adolescent suicide victims. Prostoglandins Leukot Essent Fatty Acids. 2009;80:19–26. [DOI] [PubMed] [Google Scholar]
  • 78.McNamara RK, Dwivedi Y, Roberts RC, Conley RR, Jandacek R, Rider T, Tso P, Pandey GN. Fatty acid composition of the postmortem prefrontal cortex of adolescent male and female suicide victims: comparison with young adult suicide victims with affective disorders. Meeting of the Society of Biological Psychiatry. 2009;65:S114. [DOI] [PubMed] [Google Scholar]
  • 79.Miguel-Hidalgo JJ, Baucom C, Dilley G, Overholser JC, Meltzer HY, Stockmeier CA, Rajkowska G. Glial fibrillary acidic protein immunoreactivity in the prefrontal cortex distinguishes younger from older adults in major depressive disorder. Biol Psychiatry. 2000;48:861–73. [DOI] [PubMed] [Google Scholar]
  • 80.Si X, Miguel-Hidalgo JJ, O'Dwyer G, Stockmeier CA, Rajkowska G. Age-dependent reductions in the level of glial fibrillary acidic protein in the prefrontal cortex in major depression. Neuropsychopharmacology. 2004;29:2088–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Webster MJ, Knable MB, Johnston-Wilson N, Nagata K, Inagaki M, Yolken RH. Immunohistochemical localization of phosphorylated glial fibrillary acidic protein in the prefrontal cortex and hippocampus from patients with schizophrenia, bipolar disorder, and depression. Brain Behav Immun. 2001;15:388–400. [DOI] [PubMed] [Google Scholar]
  • 82.Pellerin L, Bonvento G, Chatton JY, Pierre K, Magistretti PJ. Role of neuron-glia interaction in the regulation of brain glucose utilization. Diabetes Nutr Metab. 2002;15:268–73. [PubMed] [Google Scholar]
  • 83.Wiesinger H, Hamprecht B, Dringen R. Metabolic pathways for glucose in astrocytes. Glia. 1997;21:22–34. [DOI] [PubMed] [Google Scholar]
  • 84.Pifferi F, Roux F, Langelier B, Alessandri JM, Vancassel S, Jouin M, Lavialle M, Guesnet P. (n-3) polyunsaturated fatty acid deficiency reduces the expression of both isoforms of the brain glucose transporter GLUT1 in rats. J Nutr. 2005;135:2241–6. [DOI] [PubMed] [Google Scholar]
  • 85.Ximenes da Silva A, Lavialle F, Gendrot G, Guesnet P, Alessandri JM, Lavialle M. Glucose transport and utilization are altered in the brain of rats deficient in n-3 polyunsaturated fatty acids. J Neurochem. 2002;81:1328–37. [DOI] [PubMed] [Google Scholar]
  • 86.McNamara RK, Able J, Jandacek R, Rider T, Tso P, Lindquist DM. Perinatal n-3 fatty acid deficiency selectively reduces myo-inositol levels in the adult rat PFC: an in vivo (1)H-MRS study. J Lipid Res. 2009;50:405–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Coupland NJ, Ogilvie CJ, Hegadoren KM, Seres PC, Hanstock C, Allen PS. Decreased prefrontal myo-inositol in major depressive disorder. Biol Psychiatry. 2005;57:1526–34. [DOI] [PubMed] [Google Scholar]
  • 88.Shimon H, Agam G, Belmaker RH, Hyde TM, Kleinman JE. Reduced frontal cortex inositol levels in postmortem brain of suicide victims and patients with bipolar disorder. Am J Psychiatry. 1997;154:1148–50. [DOI] [PubMed] [Google Scholar]
  • 89.Tsukada H, Kakiuchi T, Fukumoto D, Nishiyama S, Koga K. Docosahexaenoic acid (DHA) improves the age-related impairment of the coupling mechanism between neuronal activation and functional cerebral blood flow response: a PET study in conscious monkeys. Brain Res. 2000;862:180–6. [DOI] [PubMed] [Google Scholar]
  • 90.McNamara RK, Able J, Jandacek R, Rider T, Tso P, Alfieri D, Weber W, Jarvis K, Delbello MP, et al. Docosahexaenoic acid supplementation increases prefrontal cortex activation during sustained attention in healthy boys: a placebo-controlled dose ranging fMRI study. Am J Clin Nutr. In press 2010. [DOI] [PMC free article] [PubMed]

Articles from The Journal of Nutrition are provided here courtesy of American Society for Nutrition

RESOURCES