Skip to main content

Advertisement

Log in

The Deleterious Effects of Oxidative and Nitrosative Stress on Palmitoylation, Membrane Lipid Rafts and Lipid-Based Cellular Signalling: New Drug Targets in Neuroimmune Disorders

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Oxidative and nitrosative stress (O&NS) is causatively implicated in the pathogenesis of Alzheimer’s and Parkinson’s disease, multiple sclerosis, chronic fatigue syndrome, schizophrenia and depression. Many of the consequences stemming from O&NS, including damage to proteins, lipids and DNA, are well known, whereas the effects of O&NS on lipoprotein-based cellular signalling involving palmitoylation and plasma membrane lipid rafts are less well documented. The aim of this narrative review is to discuss the mechanisms involved in lipid-based signalling, including palmitoylation, membrane/lipid raft (MLR) and n-3 polyunsaturated fatty acid (PUFA) functions, the effects of O&NS processes on these processes and their role in the abovementioned diseases. S-palmitoylation is a post-translational modification, which regulates protein trafficking and association with the plasma membrane, protein subcellular location and functions. Palmitoylation and MRLs play a key role in neuronal functions, including glutamatergic neurotransmission, and immune-inflammatory responses. Palmitoylation, MLRs and n-3 PUFAs are vulnerable to the corruptive effects of O&NS. Chronic O&NS inhibits palmitoylation and causes profound changes in lipid membrane composition, e.g. n-3 PUFA depletion, increased membrane permeability and reduced fluidity, which together lead to disorders in intracellular signal transduction, receptor dysfunction and increased neurotoxicity. Disruption of lipid-based signalling is a source of the neuroimmune disorders involved in the pathophysiology of the abovementioned diseases. n-3 PUFA supplementation is a rational therapeutic approach targeting disruptions in lipid-based signalling.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Zhao Y, Zhao B (201) Oxidative stress and the pathogenesis of Alzheimer’s disease. Oxid Med Cell Longev 2013:316523

  2. Axelsen P, Komatsu H, Murray I (2011) Oxidative stress and cell membranes in the pathogenesis of Alzheimer’s disease. Physiology 26:54–69

    Article  CAS  PubMed  Google Scholar 

  3. Hwang O (2013) Role of oxidative stress in Parkinson’s disease. Exp Neurobiol 22:11–17

    Article  PubMed  PubMed Central  Google Scholar 

  4. Pimentel C, Batista-Nascimento L, Rodrigues-Pousada C, Menezes RA (2012) Oxidative stress in Alzheimer’s and Parkinson’s diseases: insights from the yeast Saccharomyces cerevisiae. Oxid Med Cell Longev 2012:132146

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Gironi M, Borgiani B, Mariani E, Cursano C, Mendozzi L, Cavarretta R, Saresella M, Clerici M et al (2014) Oxidative stress is differentially present in multiple sclerosis courses, early evident, and unrelated to treatment. J Immunol Res 2014:1–9

    Article  CAS  Google Scholar 

  6. Ortiz G, Pacheco-Moisés F, Bitzer-Quintero O, Ramírez-Anguiano A, Flores-Alvarado L, Ramírez-Ramírez V, Macias-Islas MA, Torres-Sanchez ED (2013) Immunology and oxidative stress in multiple sclerosis: clinical and basic approach. Clin Dev Immunol 2013:1–14

    Article  CAS  Google Scholar 

  7. Morris G, Maes M (2013) Myalgic ncephalomyelitis/chronic fatigue syndrome and encephalomyelitis disseminata/multiple sclerosis show remarkable levels of similarity in phenomenology and neuroimmune characteristics. BMC Med 11:205

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kennedy G, Spence VA, McLaren M, Hill A, Underwood C, Belch JJ (2005) Oxidative stress levels are raised in chronic fatigue syndrome and are associated with clinical symptoms. Free Radic Biol Med 39:584–589

    Article  CAS  PubMed  Google Scholar 

  9. Pedrini M, Massuda R, Fries G, de Bittencourt Pasquali M, Schnorr C, Moreira J, Teixeria AL, Lobato MI et al (2012) Similarities in serum oxidative stress markers and inflammatory cytokines in patients with overt schizophrenia at early and late stages of chronicity. J Psychiatr Res 46:819–824

    Article  PubMed  Google Scholar 

  10. Kirkpatrick B, Miller BJ (2013) Inflammation and schizophrenia. Schizophr Bull 39:1174–1179

    Article  PubMed  PubMed Central  Google Scholar 

  11. Maes M, Leonard BE, Myint AM, Kubera M, Verkerk R (2011) The new ‘5-HT’ hypothesis of depression: cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry 35:702–721

    Article  CAS  PubMed  Google Scholar 

  12. Maes M, Berk M, Goehler L, Song C, Anderson G, Gałecki P, Leonard B (2012) Depression and sickness behavior are Janus-faced responses to shared inflammatory pathways. BMC Med 10:66

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Halloran M, Parakh S, Atkin JD (2013) The role of S-nitrosylation and S-glutathionylation of protein disulphide isomerase in protein misfolding and neurodegeneration. Int J Cell Biol 2013:797914

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Uchida K (2013) Redox-derived damage-associated molecular patterns: ligand function of lipid peroxidation adducts. Redox Biol 1:94–96

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Moghaddam AE, Gartlan KH, Kong L, Sattentau QJ (2011) Reactive carbonyls are a major Th2-inducing damage-associated molecular pattern generated by oxidative stress. J Immunol 187:1626–1633

    Article  CAS  PubMed  Google Scholar 

  16. Morris G, Maes M (2014) Oxidative and nitrosative stress and immune-inflammatory pathways in patients with myalgic encephalomyelitis (ME)/chronic fatigue syndrome (CFS). Curr Neuropharmacol 12:168–185

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lucas K, Maes M (2013) Role of the Toll like receptor (TLR) radical cycle in chronic inflammation: possible treatments targeting the TLR4 pathway. Mol Neurobiol 48:190–204

    Article  CAS  PubMed  Google Scholar 

  18. Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB (2010) Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 49:1603–1616

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Okamoto S, Nakamura T, Cieplak P, Chan SF, Kalashnikova E, Liao L, Saleem S, Han X et al (2014) S-nitrosylation-mediated redox transcriptional switch modulates neurogenesis and neuronal cell death. Cell Rep 8:217–228

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Nakamura T, Tu S, Akhtar MW, Sunico CR, Okamoto S, Lipton SA (2013) Aberrant protein S-nitrosylation in neurodegenerative diseases. Neuron 78:596–614

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Akhtar MW, Sunico CR, Nakamura T, Lipton SA (2012) Redox regulation of protein function via cysteine S-nitrosylation and its relevance to neurodegenerative diseases. Int J Cell Biol 2012:463756

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Wang Y, Yang J, Yi J (2012) Redox sensing by proteins: oxidative modifications on cysteines and the consequent events. Antioxid Redox Signal 16:649–657

    Article  PubMed  CAS  Google Scholar 

  23. Salaun C, Greaves J, Chamberlain L (2010) The intracellular dynamic of protein palmitoylation. J Cell Biol 191:1229–1238

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lingwood D, Simons K (2010) Lipid rafts as a membrane-organizing principle. Science 327:46–50

    Article  CAS  PubMed  Google Scholar 

  25. Yount J, Zhang M, Hang H (2013) Emerging roles for protein S-palmitoylation in immunity from chemical proteomics. Curr Opin Chem Biol 17:27–33

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ivaldi C, Martin BR, Kieffer-Jaquinod S, Chapel A, Levade T, Garin J, Journet A (2012) Proteomic analysis of S-acylated proteins in human B cells reveals palmitoylation of the immune regulators CD20 and CD23. PLoS One 7, e37187

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Henry S, Schmidt E, Fessler M, Taylor G (2014) Palmitoylation of the immunity related GTPase, Irgm1: impact on membrane localization and ability to promote mitochondrial fission. PLoS ONE 9, e95021

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Jury EC, Flores-Borja F, Kabouridis PS (2007) Lipid rafts in T cell signalling and disease. Semin Cell Dev Biol 18:608–615

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Levental I, Lingwood D, Grzybek M, Coskun U, Simons K (2010) Palmitoylation regulates raft affinity for the majority of integral raft proteins. Proc Natl Acad Sci 107:22050–22054

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Chesarino N, Hach J, Chen J, Zaro B, Rajaram M, Turner J, Schlesinger LS, Pratt MR et al (2014) Chemoproteomics reveals Toll-like receptor fatty acylation. BMC Biol 12:91

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Szabo G, Dolganiuc A, Dai Q, Pruett SB (2007) TLR4, ethanol, and lipid rafts: a new mechanism of ethanol action with implications for other receptor-mediated effects. J Immunol 178:1243–1249

    Article  CAS  PubMed  Google Scholar 

  32. Kawano Y, Kaneko-Kawano T, Shimamoto K (2014) Rho family GTPase-dependent immunity in plants and animals. Front Plant Sci 5:522

    Article  PubMed  PubMed Central  Google Scholar 

  33. Dejanovic B, Semtner M, Ebert S, Lamkemeyer T, Neuser F, Lüscher B, Meier JC, Schwarz G (2014) Palmitoylation of gephyrin controls receptor clustering and plasticity of GABAergic synapses. PLoS Biol 12, e1001908

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Björk K, Sjögren B, Svenningsson P (2010) Regulation of serotonin receptor function in the nervous system by lipid rafts and adaptor proteins. Exp Cell Res 316:1351–1356

    Article  PubMed  CAS  Google Scholar 

  35. Fiocchetti M, Ascenzi P, Marino M (2012) Neuroprotective effects of 17β-estradiol rely on estrogen receptor membrane initiated signals. Front Physiol 3:73

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Meitzen J, Luoma JI, Boulware MI, Hedges VL, Peterson BM, Tuomela K, Britson KA, Mermelstein PG (2013) Palmitoylation of estrogen receptors is essential for neuronal membrane signaling. Endocrinology 154:4293–4304

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Foster J, Vaughan R (2010) Palmitoylation controls dopamine transporter kinetics, degradation, and protein kinase C-dependent regulation. J Biol Chem 286:5175–5186

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Thomas GM, Huganir RL (2013) Palmitoylation-dependent regulation of glutamate receptors and their PDZ domain-containing partners. Biochem Soc Trans 41:72–78

    Article  CAS  PubMed  Google Scholar 

  39. Han J, Wu P, Wang F, Chen J (2015) S-palmitoylation regulates AMPA receptors trafficking and function: a novel insight into synaptic regulation and therapeutics. Acta Pharma Sin B 5:1–7

    Article  CAS  Google Scholar 

  40. Huang K, Kang MH, Askew C, Kang R, Sanders SS, Wan J, Davis NG, Hayden MR (2010) Palmitoylation and function of glial glutamate transporter-1 is reduced in the YAC128 mouse model of Huntington disease. Neurobiol Dis 40:207–215

    Article  CAS  PubMed  Google Scholar 

  41. Fröhlich M, Dejanovic B, Kashkar H, Schwarz G, Nussberger S (2014) S-palmitoylation represents a novel mechanism regulating the mitochondrial targeting of BAX and initiation of apoptosis. Cell Death Dis 5, e1057

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Bhattacharyya R, Barren C, Kovacs D (2013) Palmitoylation of amyloid precursor protein regulates amyloidogenic processing in lipid rafts. J Neurosci 33:11169–11183

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zheng B, DeRan M, Li X, Liao X, Fukata M, Wu X (2013) 2-Bromopalmitate analogues as activity-based probes to explore palmitoyl acyltransferases. J Am Chem Soc 135:7082–7085

    Article  CAS  PubMed  Google Scholar 

  44. Kong E, Peng S, Chandra G, Sarkar C, Zhang Z, Bagh M, Mukherjee A (2013) Dynamic palmitoylation links cytosol-membrane shuttling of acyl-protein thioesterase-1 and acyl-protein thioesterase-2 with that of proto-oncogene H-ras product and growth-associated protein-43. J Biol Chem 288:9112–9125

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Munday A, Lóopez J (2007) Posttranslational protein palmitoylation promoting platelet purpose. Arterioscler Thromb Vasc Biol 27:1496–1499

    Article  CAS  PubMed  Google Scholar 

  46. Aicart-Ramos C, Valero R, Rodriguez-Crespo I (2011) Protein palmitoylation and subcellular trafficking. Biochim Biophy Acta (BBA)-Biomembranes 1808:2981–2994

    Article  CAS  Google Scholar 

  47. Bijlmakers M (2008) Protein acylation and localization in T cell signaling (review). Mol Membr Biol 26:93–103

    Article  PubMed  CAS  Google Scholar 

  48. Fukata Y, Fukata M (2010) Protein palmitoylation in neuronal development and synaptic plasticity. Nat Rev Neurosci 11:161–175

    Article  CAS  PubMed  Google Scholar 

  49. Conibear E, Davis NG (2010) Palmitoylation and depalmitoylation dynamics at a glance. J Cell Sci 123:4007–4010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zheng H, Pearsall EA, Hurst DP, Zhang Y, Chu J, Zhou Y, Reggio PH, Loh HH et al (2012) Palmitoylation and membrane cholesterol stabilize μ-opioid receptor homodimerization and G protein coupling. BMC Cell Biol 13:6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. El-Husseini A-D, Bredt DS (2002) Protein palmitoylation: a regulator of neuronal development and function. Nat Rev Neurosci 3:791–802

    Article  CAS  Google Scholar 

  52. Ho GP, Selvakumar B, Mukai J, Hester LD, Wang Y, Gogos JA, Snyder SH (2011) S-nitrosylation and S-palmitoylation reciprocally regulate synaptic targeting of PSD-95. Neuron 71:131–141

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Li Y, Hu J, Höfer K, Wong AMS, Cooper JD, Birnbaum SG, Hammer RE, Hofmann SL (2010) DHHC5 interacts with PDZ domain 3 of post-synaptic density-95 (PSD-95) protein and plays a role in learning and memory. J Biol Chem 285:13022–13031

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Papoucheva E, Dumuis A, Sebben M, Richter DW, Ponimaskin EG (2004) The 5-hydroxytryptamine(1A) receptor is stably palmitoylated, and acylation is critical for communication of receptor with Gi protein. J Biol Chem 279:3280–3291

    Article  CAS  PubMed  Google Scholar 

  55. Zhang M, Wu P, Kelly F, Nurse P, Hang H (2013) Quantitative control of protein S-palmitoylation regulates meiotic entry in fission yeast. PLoS Biol 11, e1001597

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Draper J, Smith C (2010) DHHC20: a human palmitoyl acyltransferase that causes cellular transformation. Mol Membr Biol 27:123–136

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Head B, Patel H, Insel P (2014) Interaction of membrane/lipid rafts with the cytoskeleton: impact on signaling and function: membrane/lipid rafts, mediators of cytoskeletal arrangement and cell signaling. Biochim Biophys Acta (BBA)-Biomembranes 1838:532–545

    Article  CAS  Google Scholar 

  58. Goñi F (2014) The basic structure and dynamics of cell membranes: an update of the Singer Nicolson model. Biochim Biophys Acta (BBA)-Biomembranes 1838:1467–1476

    Article  CAS  Google Scholar 

  59. Bastiani M, Parton RG (2010) Caveolae at a glance. J Cell Sci 123:3831–3836

    Article  CAS  PubMed  Google Scholar 

  60. Nicolson G (2014) The fluid-mosaic model of membrane structure: still relevant to understanding the structure, function and dynamics of biological membranes after more than 40 years. Biochim Biophys Acta (BBA)-Biomembranes 1838:1451–1466

    Article  CAS  Google Scholar 

  61. Simons K, Sampaio J (2011) Membrane organization and lipid rafts. Cold Spring Harb Perspect Biol 3:004697

    Article  CAS  Google Scholar 

  62. Rushworth J, Hooper N (2011) Lipid rafts: linking Alzheimer’s amyloid-β production, aggregation, and toxicity at neuronal membranes. Int J Alzheimers Dis 2011:1–14

    Article  CAS  Google Scholar 

  63. Suzuki K (2012) Lipid rafts generate digital-like signal transduction in cell plasma membranes. Biotechnol J 7:753–761

    Article  CAS  PubMed  Google Scholar 

  64. Wang H (2014) Lipid rafts: a signaling platform linking cholesterol metabolism to synaptic deficits in autism spectrum disorders. Front Behav Neurosci 8:104

    PubMed  PubMed Central  Google Scholar 

  65. Sebastiãao A, Colino-Oliveira M, Assaife-Lopes N, Dias R, Ribeiro J (2013) Lipid rafts, synaptic transmission and plasticity: impact in age-related neurodegenerative diseases. Neuropharmacology 64:97–107

    Article  CAS  Google Scholar 

  66. Linetti A, Fratangeli A, Taverna E, Valnegri P, Francolini M, Cappello V, Matteoli M, Passafaro M et al (2010) Cholesterol reduction impairs exocytosis of synaptic vesicles. J Cell Sci 123:595–605

    Article  CAS  PubMed  Google Scholar 

  67. Boscher C, Nabi IR (2012) Caveolin-1: role in cell signaling. Adv Exp Med Biol 729:29–50

    Article  CAS  PubMed  Google Scholar 

  68. Michaela M, Michaela P (2013) Potential role of caveolin-1 in regulation of immune cell activation. Front Immunol. Conference Abstract: 15th International Congress of Immunology (ICI). doi:10.3389/conf.fimmu.2013.02.00364

  69. Chidlow J, Sessa W (2010) Caveolae, caveolins, and cavins: complex control of cellular signalling and inflammation. Cardiovasc Res 86:219–225

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Allende M, Proia RL (2002) Lubricating cell signaling pathways with gangliosides. Curr Opin Struct Biol 12:587–592

    Article  CAS  PubMed  Google Scholar 

  71. Kim O, Park E, Joe E, Jou I (2002) JAK-STAT signaling mediates gangliosides-induced inflammatory responses in brain microglial cells. J Biol Chem 277:40594–40601

    Article  CAS  PubMed  Google Scholar 

  72. Mutoh T (2013) Emergence of new roles of lipid rafts in neurological disorders. J Neurol Transl Neurosci 1:1003

    Google Scholar 

  73. Williams J, Batten S, Harris M, Rockett B, Shaikh S, Stillwell W, Wassall S (2012) Docosahexaenoic and eicosapentaenoic acids segregate differently between raft and nonraft domains. Biophys J 103:228–237

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Ruth M, Proctor S, Field C (2009) Feeding long-chain n-3 polyunsaturated fatty acids to obese leptin receptor-deficient JCR:LA-cp rats modifies immune function and lipid-raft fatty acid composition. Br J Nutr 101:1341–1350

    Article  CAS  PubMed  Google Scholar 

  75. Fan Y, McMurray D, Ly L, Chapkin R (2003) Dietary (n-3) polyunsaturated fatty acids remodel mouse T-cell lipid rafts. J Nutr 133:1913–1920

    CAS  PubMed  Google Scholar 

  76. Ma D, Seo J, Switzer K, Fan Y, McMurray D, Lupton J, Chapkin R (2004) n-3 PUFA and membrane microdomains: a new frontier in bioactive lipid research. J Nutr Biochem 15:700–706

    Article  CAS  PubMed  Google Scholar 

  77. Fan Y, Ly L, Barhoumi R, McMurray D, Chapkin R (2004) Dietary docosahexaenoic acid suppresses T cell protein kinase C theta lipid raft recruitment and IL-2 production. J Immunol 173:6151–6160

    Article  CAS  PubMed  Google Scholar 

  78. Stillwell W, Wassall S (2003) Docosahexaenoic acid: membrane properties of a unique fatty acid. Chem Phys Lipids 126:1–27

    Article  CAS  PubMed  Google Scholar 

  79. Wassall S, Stillwell W (2009) Polyunsaturated fatty acid–cholesterol interactions: domain formation in membranes. Biochim Biophys Acta (BBA)-Biomembranes 1788:24–32

    Article  CAS  Google Scholar 

  80. Shaikh S, Cherezov V, Caffrey M, Soni S, LoCascio D, Stillwell W, Wassall S (2006) Molecular organization of cholesterol in unsaturated phosphatidylethanolamines: X-ray diffraction and solid state 2H NMR reveal differences with phosphatidylcholines. J Am Chem Soc 128:5375–5383

    Article  CAS  PubMed  Google Scholar 

  81. Chapkin RS, McMurray DN, Lupton JR (2007) Colon cancer, fatty acids and anti-inflammatory compounds. Curr Opin Gastroenterol 23:48–54

    Article  CAS  PubMed  Google Scholar 

  82. Kim W, Fan Y, Barhoumi R, Smith R, McMurray D, Chapkin R (2008) n-3 polyunsaturated fatty acids suppress the localization and activation of signaling proteins at the immunological synapse in murine CD4+ T cells by affecting lipid raft formation. J Immunol 181:6236–6243

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Shaikh S, Rockett B, Salameh M, Carraway K (2009) Docosahexaenoic acid modifies the clustering and size of lipid rafts and the lateral organization and surface expression of MHC class I of EL4 cells. J Nutr 139:1632–1639

    Article  CAS  PubMed  Google Scholar 

  84. Zeyda M, Staffler G, Horejsi V, Waldhausl W, Stulnig T (2002) LAT displacement from lipid rafts as a molecular mechanism for the inhibition of T cell signaling by polyunsaturated fatty acids. J Biol Chem 277:28418–28423

    Article  CAS  PubMed  Google Scholar 

  85. Grossfield A, Feller SE, Pitman MC (2006) A role for direct interactions in the modulation of rhodopsin by omega-3 polyunsaturated lipids. Proc Natl Acad Sci U S A 103:4888–4893

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Carrillo-Tripp M, Feller SE (2005) Evidence for a mechanism by which omega-3 polyunsaturated lipids may affect membrane protein function. Biochemistry 44:10164–10169

    Article  CAS  PubMed  Google Scholar 

  87. Prior IA, Harding A, Yan J, Sluimer J, Parton RG, Hancock JF (2001) GTP-dependent segregation of H-ras from lipid rafts is required for biological activity. Nat Cell Biol 3:368–375

    Article  CAS  PubMed  Google Scholar 

  88. Eisenberg S, Shvartsman DE, Ehrlich M, Henis YI (2006) Clustering of raft-associated proteins in the external membrane leaflet modulates internal leaflet H-ras diffusion and signaling. Mol Cell Biol 26:7190–7200

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Seo J, Barhoumi R, Johnson AE, Lupton JR, Chapkin RS (2006) Docosahexaenoic acid selectively inhibits plasma membrane targeting of lipidated proteins. FASEB J 20:770–772

    CAS  PubMed  Google Scholar 

  90. Turk H, Chapkin R (2013) Membrane lipid raft organization is uniquely modified by n-3 polyunsaturated fatty acids. Prostaglandins Leukot Essent Fatty Acids 88:43–47

    Article  CAS  PubMed  Google Scholar 

  91. Greaves J, Chamberlain LH (2007) Palmitoylation-dependent protein sorting. J Cell Biol 176:249–254

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Shepherd JD, Huganir RL (2007) The cell biology of synaptic plasticity: AMPA receptor trafficking. Annu Rev Cell Dev Biol 23:613–643

    Article  CAS  PubMed  Google Scholar 

  93. Bassani S, Folci A, Zapata J, Passafaro M (2013) AMPAR trafficking in synapse maturation and plasticity. Cell Mol Life Sci 70:4411–4430

    Article  CAS  PubMed  Google Scholar 

  94. Jiang J, Suppiramaniam V, Wooten MW (2007) Posttranslational modifications and receptor-associated proteins in AMPA receptor trafficking and synaptic plasticity. Neurosignals 15:266–282

    Article  CAS  Google Scholar 

  95. Hayashi T, Rumbaugh G, Huganir RL (2005) Differential regulation of AMPA receptor subunit trafficking by palmitoylation of two distinct sites. Neuron 47:709–723

    Article  CAS  PubMed  Google Scholar 

  96. El-Husseini AE, Schnell E, Dakoji S, Sweeney N, Zhou Q, Prange O, Gauthier-Campbell C, Aguilera-Moreno A et al (2002) Synaptic strength regulated by palmitate cycling on PSD-95. Cell 108:849–863

    Article  CAS  Google Scholar 

  97. Bredt DS, Nicoll RA (2003) AMPA receptor trafficking at excitatory synapses. Neuron 40:361–379

    Article  CAS  PubMed  Google Scholar 

  98. Jackson AC, Nicoll RA (2011) Stargazing from a new vantage—TARP modulation of AMPA receptor pharmacology. J Physiol 589:5909–5910

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Yudowski GA, Olsen O, Adesnik H, Marek KW, Bredt DS (2013) Acute inactivation of PSD-95 destabilizes AMPA receptors at hippocampal synapses. PLoS One 8, e53965

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Craven SE, El-Husseini AE, Bredt DS (1999) Synaptic targeting of the postsynaptic density protein PSD-95 mediated by lipid and protein motifs. Neuron 22:497–509

    Article  CAS  PubMed  Google Scholar 

  101. Lin Y, Jover-Mengual T, Wong J, Bennett MV, Zukin RS (2006) PSD-95 and PKC converge in regulating NMDA receptor trafficking and gating. Proc Natl Acad Sci 103:19902–19907

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Cousins SL, Stephenson FA (2012) Identification of N-methyl-D-aspartic acid (NMDA) receptor subtype-specific binding sites that mediate direct interactions with scaffold protein PSD-95. J Biol Chem 287:13465–13476

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Cui H, Hayashi A, Sun H, Belmares MP, Cobey C, Phan T, Schweizer J, Salter MW et al (2007) PDZ protein interactions underlying NMDA receptor-mediated excitotoxicity and neuroprotection by PSD-95 inhibitors. J Neurosci 27:9901–9915

    Article  CAS  PubMed  Google Scholar 

  104. Hayashi T, Thomas GM, Huganir RL (2009) Dual palmitoylation of NR2 subunits regulates NMDA receptor trafficking. Neuron 64:213–226

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Iwanaga T, Tsutsumi R, Noritake J, Fukata Y, Fukata M (2009) Dynamic protein palmitoylation in cellular signaling. Prog Lipid Res 48:117–127

    Article  CAS  PubMed  Google Scholar 

  106. Nicoll RA, Tomita S, Bredt DS (2006) Auxiliary subunits assist AMPA-type glutamate receptors. Science 311:1253–1256

    Article  CAS  PubMed  Google Scholar 

  107. Noritake J, Fukata Y, Iwanaga T, Hosomi N, Tsutsumi R, Matsuda N, Tani H, Iwanari H et al (2009) Mobile DHHC palmitoylating enzyme mediates activity-sensitive synaptic targeting of PSD-95. J Cell Biol 186:147–160

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Dalva MB (2009) Neuronal activity moves protein palmitoylation into the synapse. J Cell Biol 186:7–9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Ishii H, Shibuya K, Ohta Y, Mukai H, Uchino S, Takata N, Rose JA, Kawato S (2006) Enhancement of nitric oxide production by association of nitric oxide synthase with N-methyl-D-aspartate receptors via postsynaptic density 95 in genetically engineered Chinese hamster ovary cells: real-time fluorescence imaging using nitric oxide sensitive dye. J Neurochem 96:1531–1539

    Article  CAS  PubMed  Google Scholar 

  110. Hardingham N, Dachtler J, Fox K (2013) The role of nitric oxide in pre-synaptic plasticity and homeostasis. Front Cell Neurosci 7:190

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Dyall SC, Michael-Titus AT (2008) Neurological benefits of omega-3 fatty acids. Neuromol Med 10(4):219–235, 06

    Article  CAS  Google Scholar 

  112. Delion S, Chalon S, Guilloteau D, Besnard J, Durand G (1996) Alpha-linolenic acid dietary deficiency alters age-related changes of dopaminergic and serotoninergic neurotransmission in the rat frontal cortex. J Neurochem 66:1582–1591

    Article  CAS  PubMed  Google Scholar 

  113. Zimmer L, Delpal S, Guilloteau D, Aioun J, Durand G, Chalon S (2000) Chronic n-3 polyunsaturated fatty acid deficiency alters dopamine vesicle density in the rat frontal cortex. Neurosci Lett 284:25–28

    Article  CAS  PubMed  Google Scholar 

  114. Chalon S, Delion-Vancassel S, Belzung C, Guilloteau D, Leguisquet A-M, Besnard J-C, Durand G (1998) Dietary fish oil affects monoaminergic neurotransmission and behaviour in rats. J Nutr 128:2512–2519

    CAS  PubMed  Google Scholar 

  115. Vancassel S, Leman S, Hanonick L, Denis S, Roger J, Nollet M, Bodard S, Kousignian I et al (2008) n-3 polyunsaturated fatty acid supplementation reverses stress-induced modifications on brain monoamine levels in mice. J Lipid Res 49:340–348

    Article  CAS  PubMed  Google Scholar 

  116. Kodas E, Galineau L, Bodard S, Vancassel S, Guilloteau D, Besnard J, Chalon S (2004) Serotoninergic neurotransmission is affected by n-3 polyunsaturated fatty acids in the rat. J Neurochem 89:695–702

    Article  CAS  PubMed  Google Scholar 

  117. Calon F, Lim G, Morihara T, Yang F, Ubeda O, Salem N, Frautschy SA, Cole GM (2005) Dietary n-3 polyunsaturated fatty acid depletion activates caspases and decreases NMDA receptors in the brain of a transgenic mouse model of Alzheimer’s disease. Eur J Neurosci 22:617–626

    Article  PubMed  Google Scholar 

  118. Green KN, Martinez-Coria H, Khashwji H, Hall EB, Yurko-Mauro KA, Ellis L, LaFerla FM (2007) Dietary docosahexaenoic acid and docosapentaenoic acid ameliorate amyloid-beta and tau pathology via a mechanism involving presenilin 1 levels. J Neurosci 27:4385–4395

    Article  CAS  PubMed  Google Scholar 

  119. Mirnikjoo B, Brown S, Kim H, Marangell L, Sweatt J, Weeber E (2001) Protein kinase inhibition by omega-3 fatty acids. J Biol Chem 276:10888–91086

    Article  CAS  PubMed  Google Scholar 

  120. Seung Kim HF, Weeber EJ, Sweatt DJ, Stoll AL, Marangell LB (2001) Inhibitory effects of omega-3 fatty acids on protein kinase C activity. Mol Psychiatry 6:246–248

    Article  CAS  PubMed  Google Scholar 

  121. Bourre J, Francois M, Youyou A, Dumont O, Piciotti M, Pascal G, Durand G (1989) The effects of dietary alpha-linolenic acid on the composition of nerve membranes, enzymatic activity, amplitude of electrophysiological parameters, resistance to poisons and performance of learning tasks in rats. J Nutr 119:1880–1892

    CAS  PubMed  Google Scholar 

  122. Niu S, Mitchell D, Lim S, Wen Z, Kim H, Salem N, Litman B (2004) Reduced G protein-coupled signaling efficiency in retinal rod outer segments in response to n-3 fatty acid deficiency. J Biol Chem 279:31098–31104

    Article  CAS  PubMed  Google Scholar 

  123. Kim HY (2007) Novel metabolism of docosahexaenoic acid in neural cells. J Biol Chem 282:18661–18665

    Article  CAS  PubMed  Google Scholar 

  124. Dai P, Liu X, Li Q (2012) Function of the Lck and Fyn in T cell development. Yi Chuan 34:289–295

    Article  CAS  PubMed  Google Scholar 

  125. Salmond RJ, Filby A, Qureshi I, Caserta S, Zamoyska R (2009) T-cell receptor proximal signaling via the Src-family kinases, Lck and Fyn, influences T-cell activation, differentiation, and tolerance. Immunol Rev 228:9–22

    Article  CAS  PubMed  Google Scholar 

  126. Harder T, Sangani D (2009) Plasma membrane rafts engaged in T cell signalling: new developments in an old concept. Cell Commun Signal 7:21

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Soares H, Henriques R, Sachse M, Ventimiglia L, Alonso MA, Zimmer C, Thoulouze M, Alcover A (2013) Regulated vesicle fusion generates signaling nanoterritories that control T cell activation at the immunological synapse. J Exp Med 210:2415–2433

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Balagopalan L, Ashwell BA, Bernot KM, Akpan IO, Quasba N, Barr VA, Samelson LE (2011) Enhanced T-cell signaling in cells bearing linker for activation of T-cell (LAT) molecules resistant to ubiquitylation. Proc Natl Acad Sci 108:2885–2890

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Gringhuis SI, Papendrecht-Van Der Voort EA, Leow A, Levarht E, Breedveld F, Verweij CL (2002) Effect of redox balance alterations on cellular localization of LAT and downstream T-cell receptor signaling pathways. Mol Cell Biol 22:400–411

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Ladygina N, Martin BR, Altman A (2011) Dynamic palmitoylation and the role of DHHC proteins in T cell activation and anergy. Adv Immunol 109:1–44

    Article  CAS  PubMed  Google Scholar 

  131. Hundt M, Tabata H, Jeon M, Hayashi K, Tanaka Y, Krishna R, De Giorgio L, Liu Y et al (2006) Impaired activation and localization of LAT in anergic T cells as a consequence of a selective palmitoylation defect. Immunity 24:513–522

    Article  CAS  PubMed  Google Scholar 

  132. Brownlie RJ, Zamoyska R (2013) T cell receptor signalling networks: branched, diversified and bounded. Nat Rev Immunol 13:257–269

    Article  CAS  PubMed  Google Scholar 

  133. Hrdinka M, Otahal P, Horejsi V (2012) The transmembrane region is responsible for targeting of adaptor protein LAX into “Heavy rafts”. PLoS One 7, e36330

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Filipp D, Ballek O, Manning J (2012) Lck, membrane microdomains, and TCR triggering machinery: defining the new rules of engagement. Front Immunol 3:155

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Ballek O, BrouVckov’A A, Manning J, Filipp D (2012) A specific type of membrane microdomains is involved in the maintenance and translocation of kinase active Lck to lipid rafts. Immunol Lett 142:64–74

    Article  CAS  PubMed  Google Scholar 

  136. Calder PC (2002) Dietary modification of inflammation with lipids. Proc Nutr Soc 61:345–358

    Article  CAS  PubMed  Google Scholar 

  137. Calder PC, Grimble RF (2002) Polyunsaturated fatty acids, inflammation and immunity. Eur J Clin Nutr 56(Suppl 3):S14–S19

    Article  CAS  PubMed  Google Scholar 

  138. Jump DB (2002) The biochemistry of n-3 polyunsaturated fatty acids. J Biol Chem 277:8755–8758

    Article  CAS  PubMed  Google Scholar 

  139. Gottrand F (2008) Long-chain polyunsaturated fatty acids influence the immune system of infants. J Nutr 138:1807S–1812S

    CAS  PubMed  Google Scholar 

  140. Calder P (2012) Mechanisms of action of (n-3) fatty acids. J Nutr 142:592–599

    Article  CAS  Google Scholar 

  141. Serhan CN, Hong S, Gronert K, Colgan SP, Devchand PR, Mirick G, Moussignac RL (2002) Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med 196:1025–1037

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Calder P (2006) n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr 83:1505–1519

    Google Scholar 

  143. Myers JL, Allen JC (2012) Nutrition and inflammation: insight on dietary pattern, obesity and asthma. Am J Lifestyle Med 6:14–17

    Article  Google Scholar 

  144. Rockett B, Franklin A, Harris M, Teague H, Rockett A, Shaikh S (2011) Membrane raft organization is more sensitive to disruption by (n-3) PUFA than nonraft organization in EL4 and B cells. J Nutr 141:1041–1048

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Oh DY, Lagakos WS (2011) The role of G-protein-coupled receptors in mediating the effect of fatty acids on inflammation and insulin sensitivity. Curr Opin Clin Nutr Metab Care 14:322–327

    Article  CAS  PubMed  Google Scholar 

  146. Talukdar S, Olefsky JM, Osborn O (2011) Targeting GPR120 and other fatty acid-sensing GPCRs ameliorates insulin resistance and inflammatory diseases. Trends Pharmacol Sci 32:543–550

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Shaikh S, Edidin M (2008) Polyunsaturated fatty acids and membrane organization: elucidating mechanisms to balance immunotherapy and susceptibility to infection. Chem Phys Lipids 153:24–33

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Jaudszus A, Gruen M, Watzl B, Ness C, Roth A, Lochner A, Barz D, Gabriel H et al (2013) Evaluation of suppressive and pro-resolving effects of EPA and DHA in human primary monocytes and T-helper cells. J Lipid Res 54:923–935

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Chinetti G, Fruchart JC, Staels B (2000) Peroxisome proliferator-activated receptors (PPARs): nuclear receptors at the crossroads between lipid metabolism and inflammation. Inflamm Res 49:497–505

    Article  CAS  PubMed  Google Scholar 

  150. Zúñiga J, Cancino M, Medina F, Varela P, Vargas R, Tapia G, Videla LA, Fernández V (2011) N-3 PUFA supplementation triggers PPAR-α activation and PPAR-α/NF-κB interaction: anti-inflammatory implications in liver ischemia-reperfusion injury. PLoS One 6, e28502

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Schumann J (2013) Modulation of macrophage response mechanisms against persistent pathogens by PUFA. Formatex 1799–1804

  152. Gurzell E, Teague H, Harris M, Clinthorne J, Shaikh S, Fenton J (2013) DHA-enriched fish oil targets B cell lipid microdomains and enhances ex vivo and in vivo B cell function. J Leukoc Biol 93:463–470

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Das M, Sur P, Gomes A, Vedasiromoni JR, Ganguly DK (2002) Inhibition of tumour growth and inflammation by consumption of tea. Phytother Res 16(Suppl 1):S40–S44

    Article  PubMed  Google Scholar 

  154. Verlengia R, Gorjão R, Kanunfre CC, Bordin S, Martins De Lima T, Martins EF, Curi R (2004) Comparative effects of eicosapentaenoic acid and docosahexaenoic acid on proliferation, cytokine production, and pleiotropic gene expression in Jurkat cells. J Nutr Biochem 15:657–665

    Article  CAS  PubMed  Google Scholar 

  155. Cash J, Norling L, Perretti M (2014) Resolution of inflammation: targeting GPCRs that interact with lipids and peptides. Drug Discov Today 19:1186–1192

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Anderson B, Ma D (2009) Are all n-3 polyunsaturated fatty acids created equal? Lipids Health Dis 8:33

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Das UN (2005) Can COX-2 inhibitor-induced increase in cardiovascular disease risk be modified by essential fatty acids? J Assoc Physicians India 53:623–627

    CAS  PubMed  Google Scholar 

  158. Serhan C, Savill J (2005) Resolution of inflammation: the beginning programs the end. Nat Immunol 6:1191–1197

    Article  CAS  PubMed  Google Scholar 

  159. Dietrich LE, Ungermann C (2004) On the mechanism of protein palmitoylation. EMBO Rep 5:1053–1057

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Burgoyne J, Haeussler D, Kumar V, Ji Y, Pimental D, Zee R, Costello CE, Lin C et al (2011) Oxidation of HRas cysteine thiols by metabolic stress prevents palmitoylation in vivo and contributes to endothelial cell apoptosis. FASEB J 26:832–841

    Article  PubMed  CAS  Google Scholar 

  161. Oeste CL, Díez-Dacal B, Bray F, García de Lacoba M, de la Torre BG, Andreu D, Ruiz-Sánchez AJ, Pérez-Inestrosa E et al (2011) The C-terminus of H-Ras as a target for the covalent binding of reactive compounds modulating Ras-dependent pathways. PLoS One 6, e15866

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Howie J, Swarbrick J, Shattock MJ, Fuller W (2013) Metabolic stress alters the balance between palmitoylation and glutathionylation of the Na pump regulatory protein phospholemman. Proceedings of the Physiological Society. 37th Congress of IUPS (Birmingham, UK) (2013) Proc 37th IUPS:PCD005

  163. Hess DT, Patterson SI, Smith DS, Skene JH (1993) Neuronal growth cone collapse and inhibition of protein fatty acylation by nitric oxide. Nature 366:562–565

    Article  CAS  PubMed  Google Scholar 

  164. Baker TL, Booden MA, Buss JE (2000) S-nitrosocysteine increases palmitate turnover on Ha-Ras in NIH 3T3 cells. J Biol Chem 275:22037–22047

    Article  CAS  PubMed  Google Scholar 

  165. Roy S, Plowman S, Rotblat B, Prior IA, Muncke C, Grainger S, Parton RG, Henis YI et al (2005) Individual palmitoyl residues serve distinct roles in H-ras trafficking, microlocalization, and signaling. Mol Cell Biol 25:6722–6733

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Clark K, Oelke A, Johnson M, Eilert K, Simpson P, Todd S (2004) CD81 associates with 14-3-3 in a redox-regulated palmitoylation-dependent manner. J Biol Chem 279:19401–19406

    Article  CAS  PubMed  Google Scholar 

  167. Parat M, Stachowicz R, Fox P (2002) Oxidative stress inhibits caveolin-1 palmitoylation and trafficking in endothelial cells. Biochem J 361:681–688

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Parat M, Fox PL (2004) Oxidative stress, caveolae and caveolin-1. Subcell Biochem 37:425–441

    Article  CAS  PubMed  Google Scholar 

  169. Cotticelli MG, Crabbe AM, Wilson RB, Shchepinov MS (2013) Insights into the role of oxidative stress in the pathology of Friedreich ataxia using peroxidation resistant polyunsaturated fatty acids. Redox Biol 1:398–404

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Peet M, Murphy B, Shay J, Horrobin D (1998) Depletion of omega-3 fatty acid levels in red blood cell membranes of depressive patients. Biol Psychiatry 43:315–319

    Article  CAS  PubMed  Google Scholar 

  171. Calon F, Lim GP, Yang F, Morihara T, Teter B, Ubeda O, Rostaing P, Triller A et al (2004) Docosahexaenoic acid protects from dendritic pathology in an Alzheimer’s disease mouse model. Neuron 43:633–645

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Lim K, Han C, Xu L, Isse K, Demetris AJ, Wu T (2008) Cyclooxygenase-2-derived prostaglandin E2 activates beta-catenin in human cholangiocarcinoma cells: evidence for inhibition of these signaling pathways by omega 3 polyunsaturated fatty acids. Cancer Res 68:553–560

    Article  CAS  PubMed  Google Scholar 

  173. Long EK, Murphy TC, Leiphon LJ, Watt J, Morrow JD, Milne GL, Howard JR, Picklo MJ Sr (2008) Trans-4-hydroxy-2-hexenal is a neurotoxic product of docosahexaenoic (22:6; n-3) acid oxidation. J Neurochem 105:714–724

    Article  CAS  PubMed  Google Scholar 

  174. Rosales-Corral SA, Lopez-Armas G, Cruz-Ramos J, Melnikov VG, Tan DX, Manchester LC, Munoz R, Reiter RJ (2012) Alterations in lipid levels of mitochondrial membranes induced by amyloid-β: a protective role of melatonin. Int J Alzheimers Dis 2012:459806

    PubMed  PubMed Central  Google Scholar 

  175. Véricel E, Polette A, Bacot S, Calzada C, Lagarde M (2003) Pro- and antioxidant activities of docosahexaenoic acid on human blood platelets. J Thromb Haemost 1:566–572

    Article  PubMed  Google Scholar 

  176. Pamplona R (2008) Membrane phospholipids, lipoxidative damage and molecular integrity: a causal role in aging and longevity. Biochim Biophys Acta 1777:1249–1262

    Article  CAS  PubMed  Google Scholar 

  177. Jacob RF, Mason RP (2005) Lipid peroxidation induces cholesterol domain formation in model membranes. J Biol Chem 280:39380–39387

    Article  CAS  PubMed  Google Scholar 

  178. Korytowski W, Zareba M, Girotti AW (2000) Nitric oxide inhibition of free radical-mediated cholesterol peroxidation in liposomal membranes. Biochemistry 39:6918–6928

    Article  CAS  PubMed  Google Scholar 

  179. Self-Medlin Y, Byun J, Jacob RF, Mizuno Y, Mason RP (2009) Glucose promotes membrane cholesterol crystalline domain formation by lipid peroxidation. Biochim Biophys Acta 1788:1398–1403

    Article  CAS  PubMed  Google Scholar 

  180. Mason RP, Walter MF, Day CA, Jacob RF (2006) Active metabolite of atorvastatin inhibits membrane cholesterol domain formation by an antioxidant mechanism. J Biol Chem 281:9337–9345

    Article  CAS  PubMed  Google Scholar 

  181. Yajima D, Motani H, Hayakawa M, Sato Y, Sato K, Iwase H (2009) The relationship between cell membrane damage and lipid peroxidation under the condition of hypoxia-reoxygenation: analysis of the mechanism using antioxidants and electron transport inhibitors. Cell Biochem Funct 27:338–343

    Article  CAS  PubMed  Google Scholar 

  182. De La Haba C, Palacio JR, Martínez P, Morros A (2013) Effect of oxidative stress on plasma membrane fluidity of THP-1 induced macrophages. Biochim Biophys Acta 1828:357–364

    Article  PubMed  CAS  Google Scholar 

  183. Wong-Ekkabut J, Xu Z, Triampo W, Tang IM, Tieleman DP, Monticelli L (2007) Effect of lipid peroxidation on the properties of lipid bilayers: a molecular dynamics study. Biophys J 93:4225–4236

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Venkataraman K, Khurana S, Tai TC (2013) Oxidative stress in aging—matters of the heart and mind. Int J Mol Sci 14:17897–17925

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Freikman I, Ringel I, Fibach E (2011) Oxidative stress-induced membrane shedding from RBCs is Ca flux-mediated and affects membrane lipid composition. J Membr Biol 240:73–82

    Article  CAS  PubMed  Google Scholar 

  186. Freikman I, Amer J, Cohen J, Ringel I, Fibach E (2008) Oxidative stress causes membrane phospholipid rearrangement and shedding from RBC membranes—an NMR study. Biochim Biophys Acta 1778:2388–2394

    Article  CAS  PubMed  Google Scholar 

  187. Wang H, Khor TO, Saw CL, Lin W, Wu T, Huang Y, Kong A (2010) Role of Nrf2 in suppressing LPS-induced inflammation in mouse peritoneal macrophages by polyunsaturated fatty acids docosahexaenoic acid and eicosapentaenoic acid. Mol Pharm 7:2185–2193

    Article  CAS  PubMed  Google Scholar 

  188. Ajami M, Davoodi S, Habibey R, Namazi N, Soleimani M, Pazoki-Toroudi H (2012) Effect of DHA+EPA on oxidative stress and apoptosis induced by ischemia-reperfusion in rat kidneys. Fundam Clin Pharmacol 27:593–602

    Article  PubMed  CAS  Google Scholar 

  189. Brahmbhatt V, Oliveira M, Briand M, Perrisseau G, Bastic Schmid V, Destaillats F, Pace-Asciak C, Benyacoub J et al (2013) Protective effects of dietary EPA and DHA on ischemia-reperfusion-induced intestinal stress. J Nutr Biochem 24:104–111

    Article  CAS  PubMed  Google Scholar 

  190. Martorell M, Capó X, Sureda A, Tur JA, Pons A (2014) Effects of docosahexaenoic acid diet supplementation, training, and acute exercise on oxidative balance in neutrophils. Appl Physiol Nutr Metab 39:446–457

    Article  CAS  PubMed  Google Scholar 

  191. Mathay C, Poumay Y (2010) Lipid rafts and the oxidative stress hypothesis. J Invest Dermatol 130:1457–1459

    Article  CAS  PubMed  Google Scholar 

  192. Morgan MJ, Kim YS, Liu Z (2007) Lipid rafts and oxidative stress-induced cell death. Antioxid Redox Signal 9:1471–1483

    Article  CAS  PubMed  Google Scholar 

  193. Li Q, Wang M, Tan L, Wang C, Ma J, Li N, Li Y, Xu G et al (2005) Docosahexaenoic acid changes lipid composition and interleukin-2 receptor signaling in membrane rafts. J Lipid Res 46:1904–1913

    Article  CAS  PubMed  Google Scholar 

  194. Shaikh SR (2012) Biophysical and biochemical mechanisms by which dietary N-3 polyunsaturated fatty acids from fish oil disrupt membrane lipid rafts. J Nutr Biochem 23:101–105

    Article  CAS  PubMed  Google Scholar 

  195. Zhang M, Wang S, Mao L, Leak RK, Shi Y, Zhang W, Hu X, Sun B et al (2014) Omega-3 fatty acids protect the brain against ischemic injury by activating Nrf2 and upregulating heme oxygenase 1. J Neurosci 34:1903–1915

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Nakagawa F, Morino K, Ugi S, Ishikado A, Kondo K, Sato D, Konno S, Nemoto K et al (2014) 4-Hydroxy hexenal derived from dietary n-3 polyunsaturated fatty acids induces anti-oxidative enzyme heme oxygenase-1 in multiple organs. Biochem Biophys Res Commun 443:991–996

    Article  CAS  PubMed  Google Scholar 

  197. Martín V, Fabelo N, Santpere G, Puig B, Marín R, Ferrer I, Díaz M (2010) Lipid alterations in lipid rafts from Alzheimer’s disease human brain cortex. J Alzheimers Dis 19:489–502

    PubMed  Google Scholar 

  198. Zhou X, Yang C, Liu Y, Li P, Yang H, Dai J, Qu R, Yuan L (2014) Lipid rafts participate in aberrant degradative autophagic-lysosomal pathway of amyloid-beta peptide in Alzheimer’s disease. Neural Regen Res 9:92–100

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Hicks DA, Nalivaeva NN, Turner AJ (2012) Lipid rafts and Alzheimer’s disease: protein-lipid interactions and perturbation of signaling. Front Physiol 3:189

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Williamson R, Sutherland C (2011) Neuronal membranes are key to the pathogenesis of Alzheimer’s disease: the role of both raft and non-raft membrane domains. Curr Alzheimer Res 8:213–221

    Article  CAS  PubMed  Google Scholar 

  201. Evangelisti E, Wright D, Zampagni M, Cascella R, Fiorillo C, Bagnoli S, Relini A, Nichino D et al (2013) Lipid rafts mediate amyloid-induced calcium dyshomeostasis and oxidative stress in Alzheimer’s disease. Curr Alzheimer Res 10:143–153

    Article  CAS  PubMed  Google Scholar 

  202. Oda A, Tamaoka A, Araki W (2010) Oxidative stress up-regulates presenilin 1 in lipid rafts in neuronal cells. J Neurosci Res 88:1137–1145

    CAS  PubMed  Google Scholar 

  203. Guardia-Laguarta C, Coma M, Pera M, Clarimón J, Sereno L, Agulló JM, Molina-Porcel L, Gallardo E et al (2009) Mild cholesterol depletion reduces amyloid-beta production by impairing APP trafficking to the cell surface. J Neurochem 110:220–230

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Hur JY, Teranishi Y, Kihara T, Yamamoto NG, Inoue M, Hosia W, Hashimoto M, Winblad B et al (2012) Identification of novel γ-secretase-associated proteins in detergent-resistant membranes from brain. J Biol Chem 287:11991–12005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Fernandez-Echevarria C, Díaz M, Ferrer I, Canerina-Amaro A, Marin R (2014) Aβ promotes VDAC1 channel dephosphorylation in neuronal lipid rafts. Relevance to the mechanisms of neurotoxicity in Alzheimer’s disease. Neuroscience 278:354–366

    Article  CAS  PubMed  Google Scholar 

  206. Manczak M, Reddy PH (2012) Abnormal interaction of VDAC1 with amyloid beta and phosphorylated tau causes mitochondrial dysfunction in Alzheimer’s disease. Hum Mol Genet 21:5131–5146

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Ramírez CM, González M, Díaz M, Alonso R, Ferrer I, Santpere G, Puig B, Meyer G et al (2009) VDAC and ERalpha interaction in caveolae from human cortex is altered in Alzheimer’s disease. Mol Cell Neurosci 42:172–183

    Article  PubMed  CAS  Google Scholar 

  208. Cheng JS, Dubal DB, Kim DH, Legleiter J, Cheng IH, Yu GQ, Tesseur I, Wyss-Coray T et al (2009) Collagen VI protects neurons against Abeta toxicity. Nat Neurosci 12:119–121

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Meckler X, Roseman J, Das P, Cheng H, Pei S, Keat M, Kassarjian B, Golde TE et al (2010) Reduced Alzheimer’s disease ß-amyloid deposition in transgenic mice expressing S-palmitoylation-deficient APH1aL and nicastrin. J Neurosci 30:16160–16169

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Barberger-Gateau P, Samieri C, Feart C, Plourde M (2011) Dietary omega 3 polyunsaturated fatty acids and Alzheimer’s disease: interaction with apolipoprotein E genotype. Curr Alzheimer Res 8:479–491

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Morris M, Evans D, Bienias J, Tangney C, Bennett D, Wilson R, Aggarwal N, Schneider J (2003) Consumption of fish and n-3 fatty acids and risk of incident Alzheimer disease. Arch Neurol 60:940–946

    Article  PubMed  Google Scholar 

  212. Connor W, Connor S (2007) The importance of fish and docosahexaenoic acid in Alzheimer disease. Am J Clin Nutr 85:929–930

    CAS  PubMed  Google Scholar 

  213. Fonteh A, Cipolla M, Chiang J, Arakaki X, Harrington M (2014) Human cerebrospinal fluid fatty acid levels differ between supernatant fluid and brain-derived nanoparticle fractions, and are altered in Alzheimer’s disease. PLoS One 9:100519

    Article  CAS  Google Scholar 

  214. Montine T, Morrow J (2005) Fatty acid oxidation in the pathogenesis of Alzheimer’s disease. Am J Pathol 166:1283–1289

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Markesbery W, Kryscio R, Lovell M, Morrow J (2005) Lipid peroxidation is an early event in the brain in amnestic mild cognitive impairment. Ann Neurol 58:730–735

    Article  CAS  PubMed  Google Scholar 

  216. Stoimenova A, Obreskova D, Petrov G, Peikova L, Hadjieva B, Bojkova M (2013) The application of omega-3 fatty acids in Alzheimer’s disease: the evidences from placebo-controlled clinical trials. Pharmacia 60:26–30

    CAS  Google Scholar 

  217. Cunnane S, Plourde M, Pifferi F, Bégin M, Féart C, Barberger-Gateau P (2009) Fish, docosahexaenoic acid and Alzheimer’s disease. Prog Lipid Res 48:239–256

    Article  CAS  PubMed  Google Scholar 

  218. Yurko-Mauro K, McCarthy D, Rom D, Nelson E, Ryan A, Blackwell A, Salem N Jr, Stedman M et al (2010) Beneficial effects of docosahexaenoic acid on cognition in age-related cognitive decline. Alzheimers Dement 6:456–464

    Article  CAS  PubMed  Google Scholar 

  219. Freund-Levi Y, Eriksdotter-Jonhagen M, Cederholm T, Basun H, Faxén-Irving G, Garlind A, Vedin I, Vessby B et al (2006) Omega-3 fatty acid treatment in 174 patients with mild to moderate Alzheimer disease: OmegAD study: a randomized double-blind trial. Arch Neurol 63:1402–1408

    Article  PubMed  Google Scholar 

  220. Chiu C, Su K, Cheng T, Liu H, Chang C, Dewey M, Stewart R, Huang SY (2008) The effects of omega-3 fatty acids monotherapy in Alzheimer’s disease and mild cognitive impairment: a preliminary randomized double-blind placebo-controlled study. Prog NeuroPsychopharmacol Biol Psychiatry 32:1538–1544

    Article  CAS  PubMed  Google Scholar 

  221. Shinto L, Quinn J, Montine T, Dodge HH, Woodward W, Baldauf-Wagner S, Waichunas D, Bumgarner L et al (2014) A randomized placebo-controlled pilot trial of omega-3 fatty acids and alpha lipoic acid in Alzheimer’s disease. J Alzheimers Dis 38:111–120

    CAS  PubMed  Google Scholar 

  222. Donati R, Dwivedi Y, Roberts R, Conley R, Pandey G, Rasenick M (2008) Postmortem brain tissue of depressed suicides reveals increased Gs alpha localization in lipid raft domains where it is less likely to activate adenylyl cyclase. J Neurosci 28:3042–3050

    Article  CAS  PubMed  Google Scholar 

  223. Zhang L, Rasenick M (2010) Chronic treatment with escitalopram but not R-citalopram translocates Galpha(s) from lipid raft domains and potentiates adenylyl cyclase: a 5-hydroxytryptamine transporter-independent action of this antidepressant compound. J Pharmacol Exp Ther 332:977–984

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Maes M, Bosmans E, Suy E, Vandervorst C, Dejonckheere C, Raus J (1991) Antiphospholipid, antinuclear, Epstein-Barr and cytomegalovirus antibodies, and soluble interleukin-2 receptors in depressive patients. J Affect Disord 21:133–140

    Article  CAS  PubMed  Google Scholar 

  225. Maes M, Meltzer H, Jacobs J, Suy E, Calabrese J, Minner B, Raus J (1993) Autoimmunity in depression: increased antiphospholipid autoantibodies. Acta Psychatr Scand 87:160–166

    Article  CAS  Google Scholar 

  226. Maes M, Mihaylova I, Leunis JC (2007) Increased serum IgM antibodies directed against phosphatidyl inositol (Pi) in chronic fatigue syndrome (CFS) and major depression: evidence that an IgM-mediated immune response against Pi is one factor underpinning the comorbidity between both CFS and depression. Neuro Endocrinol Lett 28:861–867

    PubMed  Google Scholar 

  227. Maes M, Kubera M, Mihaylova I, Geffard M, Galecki P, Leunis JC, Berk M (2013) Increased autoimmune responses against auto-epitopes modified by oxidative and nitrosative damage in depression: implications for the pathways to chronic depression and neuroprogression. J Affect Disord 149:23–29

    Article  CAS  PubMed  Google Scholar 

  228. Maes M, Smith R, Christophe A, Cosyns P, Desnyder R, Meltzer H (1996) Fatty acid composition in major depression: decreased omega-3 fractions in cholesteryl esters and increased C20: 4 omega 6/C20: 5 omega 3 ratio in cholesteryl esters and phospholipids. J Affect Disord 38:35–46

    Article  CAS  PubMed  Google Scholar 

  229. Maes M, Christophe A, Delanghe J, Altamura C, Neels H, Meltzer HY (1999) Lowered omega3 polyunsaturated fatty acids in serum phospholipids and cholesteryl esters of depressed patients. Psychiatry Res 85:275–291

    Article  CAS  PubMed  Google Scholar 

  230. Sublette M, Galfalvy H, Hibbeln J, Keilp J, Malone K, Oquendo M, Mann J (2014) Polyunsaturated fatty acid associations with dopaminergic indices in major depressive disorder. Int J Neuropsychopharmacol 17:383–391

    Article  CAS  PubMed  Google Scholar 

  231. Chalon S, Vancassel S, Zimmer L, Guilloteau D, Durand G (2001) Polyunsaturated fatty acids and cerebral function: focus on monoaminergic neurotransmission. Lipids 36:937–944

    Article  CAS  PubMed  Google Scholar 

  232. Grosso G, Pajak A, Marventano S, Castellano S, Galvano F, Bucolo C, Drago F, Caraci F (2014) Role of omega-3 fatty acids in the treatment of depressive disorders: a comprehensive meta-analysis of randomized clinical trials. PLoS One 9:96905

    Article  CAS  Google Scholar 

  233. Bloch M, Hannestad J (2011) Omega-3 fatty acids for the treatment of depression: systematic review and meta-analysis. Mol Psychiatry 17:1272–1282

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  234. Osher Y, Belmaker R, Nemets B (2006) Clinical trials of PUFAs in depression: state of the art. World J Biol Psychiatry 7:223–230

    Article  PubMed  Google Scholar 

  235. Martins J (2009) EPA but not DHA appears to be responsible for the efficacy of omega-3 long chain polyunsaturated fatty acid supplementation in depression: evidence from a meta-analysis of randomized controlled trials. J Am Coll Nutr 28:525–542

    Article  CAS  PubMed  Google Scholar 

  236. Stoll A, Severus W, Freeman M, Rueter S, Zboyan H, Diamond E, Cress KK, Marangell LB (1999) Omega 3 fatty acids in bipolar disorder: a preliminary double-blind, placebo-controlled trial. Arch Gen Psychiatry 56:407–412

    Article  CAS  PubMed  Google Scholar 

  237. Allen J, Halverson-Tamboli R, Rasenick M (2006) Lipid raft microdomains and neurotransmitter signalling. Nat Rev Neurosci 8:128–140

    Article  PubMed  CAS  Google Scholar 

  238. Yao J, van Kammen D (2004) Membrane phospholipids and cytokine interaction in schizophrenia. Int Rev Neurobiol 59:297–326

    Article  CAS  PubMed  Google Scholar 

  239. Condray R, Yao J (2011) Cognition, dopamine and bioactive lipids in schizophrenia. Front Biosci (Schol Ed) 3:298–330

    Google Scholar 

  240. Allen J, Yadav P, Setola V, Farrell M, Roth B (2011) Schizophrenia risk gene CAV1 is both pro-psychotic and required for atypical antipsychotic drug actions in vivo. Transcult Psychiatry 1:e33

    Article  CAS  Google Scholar 

  241. Behan AT, Byrne C, Dunn MJ, Cagney G, Cotter DR (2009) Proteomic analysis of membrane microdomain-associated proteins in the dorsolateral prefrontal cortex in schizophrenia and bipolar disorder reveals alterations in LAMP, STXBP1 and BASP1 protein expression. Mol Psychiatry 14:601–613

    Article  CAS  PubMed  Google Scholar 

  242. Berger GE, Smesny S, Amminger GP (2006) Bioactive lipids in schizophrenia. Int Rev Psychiatry 18:85–98

    Article  PubMed  Google Scholar 

  243. Emsley R, Chiliza B, Asmal L, du Plessis S, Phahladira L, van Niekerk E, van Rensburg SJ, Harvey BH (2014) A randomized, controlled trial of omega-3 fatty acids plus an antioxidant for relapse prevention after antipsychotic discontinuation in first-episode schizophrenia. Schizophr Res 158:230–235

    Article  PubMed  Google Scholar 

  244. Peet M (2008) Omega-3 polyunsaturated fatty acids in the treatment of schizophrenia. Isr J Psychiatry Relat Sci 45:19–25

    PubMed  Google Scholar 

  245. Fusar-Poli P, Berger G (2012) Eicosapentaenoic acid interventions in schizophrenia: meta-analysis of randomized, placebo-controlled studies. J Clin Psychopharmacol 32:179–185

    Article  CAS  PubMed  Google Scholar 

  246. Amminger GP, McGorry PD (2012) Update on ω-3 polyunsaturated fatty acids in early-stage psychotic disorders. Neuropsychopharmacology 37:309–310

    Article  CAS  PubMed  Google Scholar 

  247. Ellison-Wright I, Bullmore E (2009) Meta-analysis of diffusion tensor imaging studies in schizophrenia. Schizophr Res 108:3–10

    Article  PubMed  Google Scholar 

  248. Nelson B, McGorry P, Yung A, Amminger P, Francey S, Berk M, Klosterkoetter J, Schultza-Lutter F et al (2008) The NEURAPRO (North America, Europe, Australia Prodrome) Study: a multicenter RCT of treatment strategies for symptomatic patients at ultra-high risk for progression to schizophrenia and related disorders. Design and study plan. Schizophr Res 102:155

    Article  Google Scholar 

  249. McNamara RK (2011) Omega-3 fatty acid deficiency: a preventable risk factor for schizophrenia? Schizophr Res 129:215–216

    Article  PubMed  Google Scholar 

  250. Hamazaki K, Hamazaki T, Inadera H (2012) Fatty acid composition in the postmortem amygdala of patients with schizophrenia, bipolar disorder, and major depressive disorder. J Psychiatr Res 46:1024–1028

    Article  PubMed  Google Scholar 

  251. White R, Krämer-Albers EM (2014) Axon-glia interaction and membrane traffic in myelin formation. Front Cell Neurosci 7:284

    Article  PubMed  PubMed Central  Google Scholar 

  252. Krämer-Albers EM, White R (2011) From axon-glial signalling to myelination: the integrating role of oligodendroglial Fyn kinase. Cell Mol Life Sci 68:2003–2012

    Article  PubMed  CAS  Google Scholar 

  253. Liang X, Draghi NA, Resh MD (2004) Signaling from integrins to Fyn to Rho family GTPases regulates morphologic differentiation of oligodendrocytes. J Neurosci 24:7140–7149

    Article  CAS  PubMed  Google Scholar 

  254. Klein C, Kramer EM, Cardine AM, Schraven B, Brandt R, Trotter J (2002) Process outgrowth of oligodendrocytes is promoted by interaction of fyn kinase with the cytoskeletal protein tau. J Neurosci 22:698–707

    CAS  PubMed  Google Scholar 

  255. Richter-Landsberg C (2008) The cytoskeleton in oligodendrocytes. Microtubule dynamics in health and disease. J Mol Neurosci 35:55–63

    Article  CAS  PubMed  Google Scholar 

  256. Aggarwal S, Yurlova L, Snaidero N, Reetz C, Frey S, Zimmermann J, Pähler G, Janshoff A et al (2011) A size barrier limits protein diffusion at the cell surface to generate lipid-rich myelin-membrane sheets. Dev Cell 21:445–456

    Article  CAS  PubMed  Google Scholar 

  257. Schneider A, Länder H, Schulz G, Wolburg H, Nave KA, Schulz JB, Simons M (2005) Palmitoylation is a sorting determinant for transport to the myelin membrane. J Cell Sci 118:2415–2423

    Article  CAS  PubMed  Google Scholar 

  258. Pfender NA, Grosch S, Roussel G, Koch M, Trifilieff E, Greer JM (2008) Route of uptake of palmitoylated encephalitogenic peptides of myelin proteolipid protein by antigen-presenting cells: importance of the type of bond between lipid chain and peptide and relevance to autoimmunity. J Immunol 180:1398–1404

    Article  CAS  PubMed  Google Scholar 

  259. Bizzozero OA, Sanchez P, Tetzloff SU (1999) Effect of ATP depletion on the palmitoylation of myelin proteolipid protein in young and adult rats. J Neurochem 72:2610–2616

    Article  CAS  PubMed  Google Scholar 

  260. Mehta LR, Dworkin RH, Schwid SR (2009) Polyunsaturated fatty acids and their potential therapeutic role in multiple sclerosis. Nat Clin Pract Neurol 5:82–92

    Article  CAS  PubMed  Google Scholar 

  261. Torkildsen O, Wergeland S, Bakke S, Beiske AG, Bjerve KS, Hovdal H, Midgard R, Lilleås F et al (2012) ω-3 fatty acid treatment in multiple sclerosis (OFAMS study): a randomized, double-blind, placebo-controlled trial. Arch Neurol 69:1044–1051

    Article  PubMed  Google Scholar 

  262. Ramirez-Ramirez V, Macias-Islas MA, Ortiz GG, Pacheco-Moises F, Torres-Sanchez ED, Sorto-Gomez TE, Cruz-Ramos JA, Orozco-Aviña G et al (2013) Efficacy of fish oil on serum of TNF α, IL-1 β, and IL-6 oxidative stress markers in multiple sclerosis treated with interferon beta-1b. Oxid Med Cell Longev 2013:709493

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  263. Gallai V, Sarchielli P, Trequattrini A, Franceschini M, Floridi A, Firenze C, Alberti A, Di Benedetto D et al (1995) Cytokine secretion and eicosanoid production in the peripheral blood mononuclear cells of MS patients undergoing dietary supplementation with n-3 polyunsaturated fatty acids. J Neuroimmunol 56:143–153

    Article  CAS  PubMed  Google Scholar 

  264. Pantzaris MC, Loukaides GN, Ntzani EE, Patrikios IS (2013) A novel oral nutraceutical formula of omega-3 and omega-6 fatty acids with vitamins (PLP10) in relapsing remitting multiple sclerosis: a randomised, double-blind, placebo-controlled proof-of-concept clinical trial. BMJ Open 3, e002170

    Article  PubMed  PubMed Central  Google Scholar 

  265. Harbige LS, Pinto E, Xiang M, Leach M, Sharief MK (2008) PUFA in the pathogenesis and treatment of patients with multiple sclerosis. Proc Nutr Soc 67, E21

    Article  Google Scholar 

  266. Maes M, Mihaylova I, Leunis JC (2006) Chronic fatigue syndrome is accompanied by an IgM-related immune response directed against neopitopes formed by oxidative or nitrosative damage to lipids and proteins. Neuro Endocrinol Lett 27:615–621

    CAS  PubMed  Google Scholar 

  267. Maes M, Mihaylova I, Kubera M, Leunis JC, Twisk FN, Geffard M (2012) IgM-mediated autoimmune responses directed against anchorage epitopes are greater in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) than in major depression. Metab Brain Dis 27:415–423

    Article  CAS  PubMed  Google Scholar 

  268. Maes M, Mihaylova I, Leunis JC (2005) In chronic fatigue syndrome, the decreased levels of omega-3 poly-unsaturated fatty acids are related to lowered serum zinc and defects in T cell activation. Neuro Endocrinol Lett 26:745–751

    CAS  PubMed  Google Scholar 

  269. Behan PO, Behan WMH, Horrobin D (1990) Effect of high doses of essential fatty acids on the postviral fatigue syndrome. Acta Neurol Scand 1990:82209–216.216

    Google Scholar 

  270. Puri BK (2004) The use of eicosapentaenoic acid in the treatment of chronic fatigue syndrome. Prostaglandins Leukot Essent Fatty Acids 70:399–401

    Article  CAS  PubMed  Google Scholar 

  271. Cerchietti L, Navigante A, Castro M (2007) Effects of eicosapentaenoic and docosahexaenoic n-3 fatty acids from fish oil and preferential Cox-2 inhibition on systemic syndromes in patients with advanced lung cancer. Nutr Cancer 59:14–20

    Article  CAS  PubMed  Google Scholar 

  272. Alfano C, Imayama I, Neuhouser M, Kiecolt-Glaser J, Smith A, Meeske K, McTiernan A, Bernstein L et al (2012) Fatigue, inflammation, and n-3 and n-6 fatty acid intake among breast cancer survivors. J Clin Oncol 30:1280–1287

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. Hyun J, Quinn B, Madon T, Lustig S (2007) Mental health need, awareness, and use of counseling services among international graduate students. J Am Coll Health 56:109–118

    Article  PubMed  Google Scholar 

  274. Navas P, Villalba JM, de Cabo R (2007) The importance of plasma membrane coenzyme Q in aging and stress responses. Mitochondrion 7S:S34–S40

    Article  CAS  Google Scholar 

  275. Crane FL, Löw H, Sun I, Navas P, Gvozdjáková A (2014) Plasma membrane coenzyme Q: evidence for a role in autism. Biologics 8:199–205

    CAS  PubMed  PubMed Central  Google Scholar 

  276. Morris G, Anderson G, Berk M, Maes M (2013) Coenzyme Q10 depletion in medical and neuropsychiatric disorders: potential repercussions and therapeutic implications. Mol Neurobiol 48:883–903

    Article  CAS  PubMed  Google Scholar 

  277. Noh YH, Kim KY, Shim MS, Choi SH, Choi S, Ellisman MH, Weinreb RN, Perkins GA et al (2013) Inhibition of oxidative stress by coenzyme Q10 increases mitochondrial mass and improves bioenergetic function in optic nerve head astrocytes. Cell Death Dis 4, e820

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  278. Ochoa JJ, Quiles JL, Huertas JR, Mataix J (2005) Coenzyme Q10 protects from aging-related oxidative stress and improves mitochondrial function in heart of rats fed a polyunsaturated fatty acid (PUFA)-rich diet. J Gerontol A Biol Sci Med Sci 60:970–975

    Article  PubMed  Google Scholar 

  279. Hargreaves IP (2014) Coenzyme Q10 as a therapy for mitochondrial disease. Int J Biochem Cell Biol 49:105–111

    Article  CAS  PubMed  Google Scholar 

  280. Miyamae T, Seki M, Naga T, Uchino S, Asazuma H, Yoshida T, Iizuka Y, Kikuchi M et al (2013) Increased oxidative stress and coenzyme Q10 deficiency in juvenile fibromyalgia: amelioration of hypercholesterolemia and fatigue by ubiquinol-10 supplementation. Redox Rep 18:12–19

    Article  CAS  PubMed  Google Scholar 

  281. Sanoobar M, Eghtesadi S, Azimi A, Khalili M, Jazayeri S, Reza Gohari M (2013) Coenzyme Q10 supplementation reduces oxidative stress and increases antioxidant enzyme activity in patients with relapsing-remitting multiple sclerosis. Int J Neurosci 123:776–782

    Article  CAS  PubMed  Google Scholar 

  282. Lee BJ, Tseng YF, Yen CH, Lin PT (2013) Effects of coenzyme Q10 supplementation (300 mg/day) on antioxidation and anti-inflammation in coronary artery disease patients during statins therapy: a randomized, placebo-controlled trial. Nutr J 12:142

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  283. Wagner AE, Ernst IM, Birringer M, Sancak O, Barella L, Rimbach G (2012) A combination of lipoic acid plus coenzyme Q10 induces PGC1α, a master switch of energy metabolism, improves stress response, and increases cellular glutathione levels in cultured C2C12 skeletal muscle cells. Oxid Med Cell Longev 2012:835970

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  284. Carlson DA, Smith AR, Fischer SJ, Young KL, Packer L (2007) The plasma pharmacokinetics of R-(+)-lipoic acid administered as sodium R-(+)-lipoate to healthy human subjects. Altern Med Rev 12:343–351

    PubMed  Google Scholar 

  285. Packer L, Tritschler HJ, Wessel K (1997) Neuroprotection by the metabolic antioxidant alpha-lipoic acid. Free Radic Biol Med 22:359–378

    Article  CAS  PubMed  Google Scholar 

  286. Li DW, Li GR, Lu Y, Liu ZQ, Chang M, Yao M, Cheng W, Hu LS (2013) Alpha-lipoic acid protects dopaminergic neurons against MPP+-induced apoptosis by attenuating reactive oxygen species formation. Int J Mol Med 32:108–114

    CAS  PubMed  Google Scholar 

  287. Sudheesh NP, Ajith TA, Janardhanan KK (2013) Hepatoprotective effects of DL-alpha-lipoic acid and alpha-tocopherol through amelioration of the mitochondrial oxidative stress in acetaminophen challenged rats. Toxicol Mech Methods 23:368–376

    Article  CAS  PubMed  Google Scholar 

  288. Dean O, Giorlando F, Berk M (2011) N-acetylcysteine in psychiatry: current therapeutic evidence and potential mechanisms of action. J Psychiatry Neurosci 36:78–86

    Article  PubMed  PubMed Central  Google Scholar 

  289. Berk M, Malhi GS, Gray LJ, Dean OM (2013) The promise of N-acetylcysteine in neuropsychiatry. Trends Pharmacol Sci 34:167–177

    Article  CAS  PubMed  Google Scholar 

  290. Morris G, Anderson G, Dean O, Berk M, Galecki P, Martin-Subero M, Maes M (2014) The glutathione system: a new drug target in neuroimmune disorders. Mol Neurobiol 50:1059–1084

    Article  CAS  PubMed  Google Scholar 

  291. Vaughan RA, Foster JD (2013) Mechanisms of dopamine transporter regulation in normal and disease states. Trends Pharmacol Sci 34:489–496

    Article  CAS  PubMed  Google Scholar 

  292. Cherukuri A, Carter RH, Brooks S, Bornmann W, Finn R, Dowd CS, Pierce SK (2004) B cell signaling is regulated by induced palmitoylation of CD81. J Biol Chem 279:31973–31982

    Article  CAS  PubMed  Google Scholar 

  293. Poggi M, Kara I, Brunel J, Landrier J, Govers R, Bonardo B, Fluhrer R, Haass C et al (2013) Palmitoylation of TNF alpha is involved in the regulation of TNF receptor 1 signalling. Biochim Biophys Acta 1833:602–612

    Article  CAS  PubMed  Google Scholar 

  294. Yount JS, Moltedo B, Yang YY, Charron G, Moran TM, López CB, Hang HC (2010) Palmitoylome profiling reveals S-palmitoylation-dependent antiviral activity of IFITM3. Nat Chem Biol 6:610–614

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  295. Meiler S, Baumer Y, Huang Z, Hoffmann FW, Fredericks GJ, Rose AH, Norton RL, Hoffmann PR et al (2013) Selenoprotein K is required for palmitoylation of CD36 in macrophages: implications in foam cell formation and atherogenesis. J Leukoc Biol 93:771–780

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  296. Ren W, Jhala U, Du K (2013) Proteomic analysis of protein palmitoylation in adipocytes. Adipocyte 2:17–27

    Article  PubMed  PubMed Central  Google Scholar 

  297. Wei X, Yang Z, Rey F, Ridaura V, Davidson N, Gordon J, Semenkovich C (2012) Fatty acid synthase modulates intestinal barrier function through palmitoylation of mucin 2. Cell Host Microbe 11:140–152

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  298. Sánchez-Wandelmer J, Dávalos A, Herrera E, Giera M, Cano S, de la Peña G, Lasunción MA, Busto R (2009) Inhibition of cholesterol biosynthesis disrupts lipid raft/caveolae and affects insulin receptor activation in 3T3-L1 preadipocytes. Biochim Biophys Acta 1788:1731–1739

    Article  PubMed  CAS  Google Scholar 

  299. Chakrabandhu K, Hérincs Z, Huault S, Dost B, Peng L, Conchonaud F, Marguet D, He HT et al (2006) Palmitoylation is required for efficient Fas cell death signaling. EMBO J 26:209–220

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  300. He B, Zhang Y, Richardson M, Zhang J, Rubinstein E, Zhang X (2011) Differential functions of phospholipid binding and palmitoylation of tumour suppressor EWI2/PGRL. Biochem J 437:399–411

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  301. Konitsiotis A, Chang S, Jovanović B, Ciepla P, Masumoto N, Palmer CP, Tate EW, Couchman JR et al (2014) Attenuation of hedgehog acyltransferase-catalyzed sonic hedgehog palmitoylation causes reduced signaling, proliferation and invasiveness of human carcinoma cells. PLoS ONE 9, e89899

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  302. Babina I, McSherry E, Donatello S, Hill A, Hopkins A (2014) A novel mechanism of regulating breast cancer cell migration via palmitoylation-dependent alterations in the lipid raft affiliation of CD44. Breast Cancer Res 161:R19

    Article  CAS  Google Scholar 

  303. Murai T (2012) The role of lipid rafts in cancer cell adhesion and migration. Int J Cell Biol 2012:1–6

    Article  CAS  Google Scholar 

  304. Takahashi T, Suzuki T (2011) Function of membrane rafts in viral lifecycles and host cellular response. Biochem Res Int 2011:245090

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  305. Blanc M, Blaskovic S, van der Goot F (2013) Palmitoylation, pathogens and their host. Biochem Soc Trans 41:84–88

    Article  CAS  PubMed  Google Scholar 

  306. Zaas D, Duncan M, Rae Wright J, Abraham S (2005) The role of lipid rafts in the pathogenesis of bacterial infections. Biochim Biophys Acta 1746:305–313

    Article  CAS  PubMed  Google Scholar 

  307. Demers A, Ran Z, Deng Q, Wang D, Edman B, Lu W, Li F (2014) Palmitoylation is required for intracellular trafficking of influenza B virus NB protein and efficient influenza B virus growth in vitro. J Gen Virol 95:1211–1220

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

The authors would like to express their thanks to Victoria Storey for her invaluable secretarial services. No specific funding was obtained for this specific review.

Competing Interests

The authors declare that they have no competing interests.

Authors’ Contributions

GM and MM participated in the design of this review, while KW, BKP and MB helped to draft the paper. All authors read and approved the final version of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michael Maes.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Morris, G., Walder, K., Puri, B.K. et al. The Deleterious Effects of Oxidative and Nitrosative Stress on Palmitoylation, Membrane Lipid Rafts and Lipid-Based Cellular Signalling: New Drug Targets in Neuroimmune Disorders. Mol Neurobiol 53, 4638–4658 (2016). https://doi.org/10.1007/s12035-015-9392-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-015-9392-y

Keywords

Navigation