Hostname: page-component-8448b6f56d-c4f8m Total loading time: 0 Render date: 2024-04-17T04:13:58.682Z Has data issue: false hasContentIssue false

Polyunsaturated fatty acids in the pathogenesis and treatment of multiple sclerosis

Published online by Cambridge University Press:  01 October 2007

Laurence S. Harbige*
Affiliation:
Centre for Bioscience Research, School of Science, University of Greenwich at Medway, United Kingdom Medway School of Pharmacy, University of Kent and University of Greenwich, United Kingdom
Mohammad K. Sharief
Affiliation:
Department of Neurology, King's, Guy's and St Thomas' Hospital, London, United Kingdom
*
*Corresponding author: Dr Laurence Harbige, fax 0044 (0)1813319805, email L.Harbige@gre.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Epidemiological, biochemical, animal model and clinical trial data described in this overview strongly suggest that polyunsaturated fatty acids, particularly n-6 fatty acids, have a role in the pathogenesis and treatment of multiple sclerosis (MS). Data presented provides further evidence for a disturbance in n-6 fatty acid metabolism in MS. Disturbance of n-6 fatty acid metabolism and dysregulation of cytokines are shown to be linked and a “proof of concept clinical trial” further supports such a hypothesis. In a randomised double-blind, placebo controlled trial of a high dose and low dose selected GLA (18 : 3n-6)-rich oil and placebo control, the high dose had a marked clinical effect in relapsing-remitting MS, significantly decreasing the relapse rate and the progression of disease. Laboratory findings paralleled clinical changes in the placebo group in that production of mononuclear cell pro-inflammatory cytokines (TNF-α, IL-1β) was increased and anti-inflammatory TGF-β markedly decreased with loss of membrane n-6 fatty acids linoleic (18 : 2n-6) and arachidonic acids (20 : 4n-6). In contrast there were no such changes in the high dose group. The improvement in disability (Expanded Disability Status Scale) in the high dose suggests there maybe a beneficial effect on neuronal lipids and neural function in MS. Thus disturbed n-6 fatty acid metabolism in MS gives rise to loss of membrane long chain n-6 fatty acids and loss of the anti-inflammatory regulatory cytokine TGF-β, particularly during the relapse phase, as well as loss of these important neural fatty acids for CNS structure and function and consequent long term neurological deficit in MS.

Type
Full Papers
Copyright
Copyright © The Authors 2007

Background to multiple sclerosis and its pathogenesis

Multiple sclerosis (MS) is a CNS-specific demyelinating disease, and is the most common neurological disorder that occurs in young adultsReference Ewing and Bernard1, Reference Noseworthy2. The majority of patients with MS have the relapsing-remitting form of the disease, characterised by attacks (relapses) interspersed with periods of recovery (remission). The disease is most prevalent (30–100+ cases per 100 000 people) in Western Europe, Southern Canada, Northern United States, Southern Australia and New Zealand and of low frequency (0–19 per 100,000) in Asia, Central America, Africa and Greenland (See Fig. 1). Between 2 and 3 million people Worldwide are thought to live with MS. Although the aetiology of MS remains unknown there is strong evidence for the presence of autoimmune mechanisms in the disease pathogenesisReference Martino and Hartung3, Reference Hafler4. Studies have shown that MS patients have a much higher number of neuroantigen e.g. myelin basic protein (MBP) and myelin oligodendrocyte glycoprotein (MOG) autoreactive T-cells, which are in an increased state of activation compared with healthy controls, and which increase during exacerbationReference Hafler4Reference Bielekova, Goodwin, Richert, Cortese, Kondo, Afshar, Gran and Eaton13.

Fig. 1 Geographical distribution of multiple sclerosis (from Adams C (1989) colour atlas of multiple sclerosis and other myelin disorders, wolfe medical publications ltd, with permission).

Cytokines from activated T cells and macrophages have been strongly implicated in the pathogenesis of MSReference Navikas and Link14. For example, the up-regulation of adhesion molecules on endothelial cells and the subsequent infiltration of activated T cells into the CNS are immunopathogenic events controlled by pro-inflammatory cytokines such as tumour necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interferon-γ (IFN-γ)Reference McCarron, Wang and Racke15. Furthermore studies have shown that these cytokines exert direct myelinotoxic propertiesReference Selmaj, Raine and Farooq16, Reference Vartanian, Li and Zhao17 and prolong the disease process in experimental autoimmune encephalomyelitis (EAE), an animal model of MSReference Kuroda and Shimamoto18, Reference Issazadeh, Lorentzen and Mustafa19, Reference Ruddle, Bergman and McGrath20. TNF-α, IL-1β and IFN-γ have all been shown to be present in CNS active lesions in MS and elevated amounts of these cytokines are secreted from MS peripheral blood mononuclear cells (PBMC)Reference Cannella and Raine21Reference Hollifield, Harbige, Pham-Dinh and Sharief25. Many studies, including our own, have also shown that an increase in these inflammatory cytokines coincides with the relapse phase of the diseaseReference Hollifield, Harbige, Pham-Dinh and Sharief25Reference Lu, Jensen and Arnason33. Furthermore some studies have shown that transforming growth factor-beta1 (TGF-β1), a potent anti-inflammatory and immunosuppressive cytokine, is reduced during the relapse phase and increases as the patient enters remissionReference Hollifield, Harbige, Pham-Dinh and Sharief25, Reference Bertolotto, Capobianco and Malucchi34, Reference Mokhtarian, Shi and Shirazian35. In addition we have demonstrated that the balance between biologically active TGF-β1 and the pro-inflammatory TNF-α, IL-1β and IFN-γ is dysregulated during MS relapse-remissionReference Hollifield, Harbige, Pham-Dinh and Sharief25. The actual processes of axonal damage e.g. chronic inflammation, demyelination and astrogliosis in MS is complex but white matter inflammation and demyelination are considered to determine disease severity, whilst recent studies suggest that axonal damage in MS begins in the early stages of the disease and contributes to disabilityReference De Stefano, Narayanan, Francis, Arnaoutelis, Tartaglia, Antel, Matthews and Arnold36, Reference Bjartmar, Wujek and Trapp37. Furthermore some have considered metabolic disturbances in some MS patients to be behind primary oligodendrocyte damage with secondary autoimmune-demyelinationReference Lassmann, Bruck and Lucchinettii38, Reference Matute and Perez-Cerda39.

Nutritional epidemiology of multiple sclerosis

Over half a century ago Roy SwankReference Swank40 found a positive relationship between fat intake as well as annual milk production and MS in Scandinavian countries. Furthermore studies by Alter et al.Reference Alter, Yamoor and Harshe41 implicated animal fat rich in saturated fatty acids as a causal factor in MS and WolfgramReference Wolfgram42 in an analysis of World Health Organisation (WHO) annual mortality statistics found a similar geographical distribution between MS coronary heart disease and cancer of the colon. In the multivariate analysis (inclusive of socioeconomic and medical services) of 20 countries Arganoff and GoldbergReference Agranoff and Goldberg43 not only implicated meat and dairy fats in positive correlations with MS, as noted previously, but also vegetable, seed, nut and fish, foods rich in both the n-6 and n-3 polyunsaturated fatty acids, in negative correlations with MS. Several other studies also confirmed strong MS associations with dairy and other animal fatsReference Ghadirian, Jain and Ducic44Reference Malosse, Perron, Sasco and Seigneurin46. Similar observations have also been made more recently by Esparsa et al. Reference Esparza, Sasaki and Kesteloot47 in a large study (36 countries) assessing the impact of diet on MS mortality. They found that the higher the saturated fatty acid intake the higher the MS mortality and the higher the polyunsaturated to saturated fatty acid ratio the lower the MS mortality. However the large single country study, the Nurses' Health Study in the USA failed to show any relationship between MS and fat intake in womenReference Zhang, Willet, Hernan, Olek and Ascherio48. Thus the majority of epidemiological studies indicate that foods rich in saturated fatty acids are detrimentally associated with MS, whilst polyunsaturated fatty acid rich foods are beneficially associated with MS.

Biochemical and metabolic studies of fatty acids in multiple sclerosis

There is much evidence that the n-6 fatty acids particularly linoleic (18 : 2n-6) and arachidonic acids (20 : 4n-6) are reduced in the plasma, platelets, erythrocytes, leucocytes and cerebrospinal fluid with changes in the unsaturated fatty acid composition of brain white matter in MS patients, much of this early work being undertaken at the National Hospital, Queen Square in LondonReference Baker, Thompson and Zilkha49Reference Holman, Johnson and Kokmen60. But there are also inconsistent reportsReference Shukla and Clausen61Reference Nightingale, Woo and Smith64and Love et al.Reference Love, Cashell, Reynolds and Callaghan65 observed that reduced linoleic acid was not specific to MS and occurred in patients with acute non-neurological illness. However many of these differences between studies are perhaps not surprising given cultural and ethnic differences, dietary variability (particularly when someone is ill), possible desaturase gene polymorphism, disease variability, serum verses cellular fatty acids and methodological differences for example total lipid fatty acids verses phospholipids fatty acid analysis.

Previously we proposed nervonic acid as a marker of CNS myelin damage in MSReference Jones, Harbige, Clifford Rose and Jones66 and found that MS patients consuming a diet rich in polyunsaturated fatty acids particularly linoleic acid had an inverse relationship between erythrocyte membrane linoleic acid and nervonic acid (24 : 1)Reference Harbige, Crawford, Jones, Preece and Forti67. A similar finding was described by Homa et al. Reference Homa, Conroy, Belin, Smith, Monro and Zilkha68, showing a decrease in erythrocyte lignoceric acid (24 : 0) in sunflower oil (rich in linoleic acid) supplemented MS patients. In an open, uncontrolled 2 year fish oil intervention study by Nordvik et al.Reference Nordvik, Myhr, Nyland and Bjerve69 in MS, they observed significant reductions in plasma total phospholipid nervonic and lignoceric acids with time and clinical improvement. Taken together the above indicate that nervonic and lignoceric acids could be useful pathogenic biomarkers of myelin damage and/or biomarkers for monitoring fatty acid treatments. We also found that the atypical erythrocyte electrophoretic response of MS patients was positively correlated with membrane linoleic acid and could be corrected by a diet rich in polyunsaturated fatty acids particularly linoleic acidReference Harbige, Crawford, Jones, Preece and Forti67. This is in agreement with Field and JoyceReference Field, Joyce and Smith71 who found an increase in erythrocyte electrophoretic response in MS patients supplemented with evening primrose oil (EPO). However, Field et al.Reference Field, Joyce and Smith71, Field and JoyceReference Field and Joyce70 interpreted their electrophoretic results, without an analysis of membrane fatty acids, as an effect of the gamma-linolenic (GLA, 18 : 3n-6) component of the oil. EPO contains about 70 % linoleic acid and 8–10 % GLA, therefore it is more likely that the effect observed by Field et al was due to the linoleic acid component of the oil rather than the GLA.

We have also investigated the metabolic relationships between the n-6 fatty acids in both healthy controls and MS PBMC total phospholipids (Figs. 2, 3 and 4). Both controls and MS patients (remission phase) demonstrate a positive correlation between linoleic acid (18 : 2n-6) and arachidonic acid (20 : 4n-6) as expected, although these n-6 fatty acids were low in a proportion of the MS patients studied (Fig. 2). Moreover, the relationship between linoleic acid (18 : 2n-6) and dihomo-γ-linolenic acid (DGLA), and also between DGLA (20 : 3n-6) and arachidonic acid (20 : 4n-6) is clearly disturbed in MS compared with healthy controls (Figs. 3 and 4). This may indicate a problem with Δ6 and Δ5 desaturation and / or a greater requirement for these n-6 fatty acids in many of the MS patients studied, about 20–30 percent of the patients showed lower than normal PBMC phospholipid DGLA and arachidonic acid. In agreement with our findings Homa et al. Reference Homa, Belin and Smith55 has also reported a similar disturbance in the relationship between linoleic acid and arachidonic acid in MS erythrocyte membrane lipids compared to healthy controls. Furthermore when we compared MS and healthy control PBMC total phospholipid 20 : 2n-6 we found a significant 2 fold higher 20 : 2n-6 in MS patients in remission compared to healthy controls and a significant 4 fold higher 20 : 2n-6 in the relapse phase of the disease. It appears that in MS there is a very active elongation of 18 : 2n-6 to 20 : 2n-6 in PBMCs and that this is even higher in the relapse phase (accounting for the low 18 : 2n-6) indicating a disturbance in the normal metabolism or a higher requirement for DGLA and arachidonic acid (20 : 2n-6 maybe further Δ8 desaturated to DGLA?) by these n-6 fatty acid (20 : 4n-6) rich cellsReference Harbige72, or both. This may also be reflective of the demand of cells and myelin in the brain which are also n-6 fatty acid-rich (20 : 4n-6 and 22 : 4n-6), significantly Stanley Rapoport of the NIH has shown that the human brain requires 4 times the amount of arachidonic acid (20 : 4n-6) than docosahexaenoic acid (22 : 6n-3) on a daily bases (ISSFAL 2006). In relation to 20 : 2n-6, although not discussed by the authors, the Nordvik et al.Reference Nordvik, Myhr, Nyland and Bjerve69 study demonstrated a reduction with time of 20 : 2n-6 running parallel with clinical improvement, a similar finding to the nervonic and lignoceric acids mentioned earlier. Therefore 20 : 2n-6 may also be a useful marker of disease progression and/or monitoring fatty acid treatments in MS.

Fig. 2 Relationship between linoleic acid (18 : 2n-6) and arachidonic acid (20 : 4n-6) in peripheral blood mononuclear cell total phospholipids of healthy controls and multiple sclerosis.

Fig. 3 Relationship between linoleic acid (18 : 2n-6) and dihomo-γ-linolenic acid (20 : 3n-6) in peripheral blood mononuclear cell total phospholipids of healthy controls and multiple sclerosis.

Fig. 4 Relationship between dihomo-γ-linolenic acid (20 : 3n-6) and arachidonic acid (20 : 4n-6) in peripheral blood mononuclear cell total phospholipids of healthy controls and multiple sclerosis.

Fatty acids and animal models of multiple sclerosis

Experimental autoimmune encephalomyelitis (EAE) is an experimentally induced CD4+T cell mediated autoimmune-inflammatory and deyelinating disease in rodents often used as an animal model of MS. Studies in guinea pig and rat EAE treated with linoleic acid alone or a high linoleic and low γ-linolenic (GLA) acid rich oil (ratio 7 : 1) respectively, have shown partial suppression of the incidence and severity of EAEReference Meade, Mertin, Sheena and Hunt73, Reference Mertin and Stackpoole74. In a series of experiments we demonstrated important disease modifying effects of linoleic acid-rich oil (containing no GLA) and GLA-rich oil on clinical and histopathological manifestations of EAE. Depending on dose GLA was completely protective in EAE, whereas linoleic acid had a dose dependent action on the clinical severity of EAE, although not abolishing itReference Harbige, Yeatman, Amor and Crawford75, Reference Harbige, Layward and Morris-Downes76. Natural recovery in EAE is mediated by expansion of suppressor lymphoid cellsReference Adda, Beraud and Depieds77 some of which have been characterised as TGF-β producing CD4+T cells by Karpus and SwanborgReference Karpus and Swanborg78. Furthermore administration of TGF-β protects in acute and relapsing EAEReference Rack, Sriram, Calrlini, Cannella, Raine and McFarlin79, Reference Santambrogio, Hochwald, Saxena, Leu, Martz, Carlino, Ruddle, Palladino, Gold and Thorbecke80 and prostaglandin inhibitors such as indomethacin augment EAEReference Ovadia and Paterson81. In-addition during the natural recovery phase from EAE TGF-β secreting T cells can inhibit EAE effector cells and TGF-β is expressed in the CNSReference Karpus and Swanborg78, Reference Liblau, Singer and McDevitt82, Reference Khoury, Hancock and Weiner83. Consistent with these findings the protective effect of GLA-rich oil in EAE is linked to increased T cell TGF-β transcription and increased production of PGE2Reference Harbige, Layward and Morris-Downes76.

Clinical trials and intervention studies in multiple sclerosis with fatty acids

Clinical trials to test the efficacy of linoleic acid-rich sunflower oil in MS patients by Miller et al. Reference Bates, Fawcett, Shaw and Weightman84 and Bates et al. Reference Millar, Zilkha, Langman and Payling-Wright85 over 2 years showed a reduction in the relapse rate and severity of the disease relapse, but Paty et al. Reference Paty, Cousin, Read and Adlakha86 found no such effect. Nevertheless, Dworkin et al. Reference Dworkin, Bates and Millar87 in a statistical revaluation of the combined data of all three trials revealed significantly reduced relapse rate and severity, and in mildly affected a decrease in the long term progression of the disease. The Millar et al. Reference Bates, Fawcett, Shaw and Weightman84 study based in two centres London and Belfast is particularly interesting as they observed that “the severity of the relapses, differed markedly between the treated and the control groups at both centres” relapses being twice as severe in the control group. Compared with current β-interferon treatment of MS the efficacy of linoleic acid-rich sunflower oil in the Miller et al study is quite remarkable. Fish oil rich in long chain n-3 fatty acids has also been studied in MSReference Bates, Cartlidge and French88, no significant differences between fish oil treated and untreated MS patients was observed, there was, however, a trend for less deterioration in the fish oil treated group. In a 2 year open intervention study MS patients given fish oil and advised to lower their saturated fat intake had significant reductions in the relapse rate and disability progression as measured by the Expanded Disability Status Scale (EDSS)Reference Nordvik, Myhr, Nyland and Bjerve69 which quantifies disability in MS in eight functional systems (pyramidal, cerebellar, brainstem, sensory, bowel and bladder, visual, cerebral, other). It also appears based on open studies by us and by Roy Swank that long term low saturated fat diets containing both n-6 and n-3 fatty acids improve the course of the diseaseReference Swank89Reference Harbige, Jones, Jenkins, Fitzgerald, Forti and Budowski93.

Based on our MS fatty acid metabolic data and experimental animal model work, described above, we undertook a randomised double-blind placebo controlled trial to determine the effects of supplementation with a selected GLA (18 : 3n-6)-rich borage oil. This oil, BGC20-884 was high in sn-2 GLA, low in monoenes and contained only natural levels of vitamin E. This study evaluated two doses of BGC20-884 (low dose - 5 gram and high dose - 14 gram per day) and a placebo control (polyethylene glycol 400) on the clinical course and PBMC cytokine and membrane fatty acid profiles of 36 patients with active MS over 18 monthsReference Harbige, Hollifield, Pinto, Xiang, Leach and Sharief94. Patients were diagnosed and assessed using international criteria for MS. Relapse rate and EDSS (Expanded Disability Status Scale) were assessed every three months and blood taken and PBMCs isolated for cytokine studies and membrane fatty acids. High dose BGC20-884 treatment markedly and significantly reduced the relapse rate (Fig. 5) and disability progression as measured by EDSS (Fig. 6) compared with the placebo control and low dose BGC20-884 treatment. In patients where we had follow up samples available PBMC cytokine changes were found to run parallel with the clinical findings e.g. the placebo control group showed significant decreases in the TGF-β/TNF-α and TGF-β/IL-1β ratios and associated loss of n-6 fatty acids particularly linoleic (18 : 2n-6) and arachidonic acid (20 : 4n-6) over time. Consistent with our findings Navarro and SeguraReference Navarro and Segura59 also found significant loss of linoleic and arachidonic acids over time in MS erythrocyte phospholipids. In contrast high dose BGC20-884 treatment showed no changes in TGF-β/TNF-α and TGF-β/IL-1β ratios and no changes in membrane n-6 fatty acids compared with the placebo group. We also found positive correlations between PBMC phospholipid arachidonic acid composition and TGF-β1 production (r = 0·26, P < 0·02, n = 73) and DGLA and TGF-β1 production (r = 0·36, P < 0·001, n = 74) ex vivo when all samples were included in the analysis. The EDSS improvement in the high dose group also suggests there maybe a beneficial effect on neuronal lipids and neural function in MS. The study thus further supports our hypothesis of dysregulation of fatty acid metabolism and cytokines in MSReference Hollifield, Harbige, Pham-Dinh and Sharief25, Reference Harbige, Layward and Morris-Downes76.

Fig. 5 Mean annualised relapse rate per patient for multiple sclerosis patients receiving high (n = 11) and low dose (n = 7) GLA-rich oil or placebo control (n = 10) over 18 months.

Fig. 6 Disability progression as measured by the EDSS (Expanded Disability Status Scale) in multiple sclerosis patients receiving high (n = 11) and low dose (n = 7) GLA-rich oil or placebo control (n = 10) over 18 months.

To summarise and extend this section on clinical trials there is evidence to show a beneficial effect of n-6 and possibly n-3 fatty acids in MS. The mechanisms by which the n-3 or the n-6 fatty acids influence the immune-inflammatory response in MS are however likely to be differentReference Harbige72. Both the n-6 (arachidonic acid) and n-3 (docosahexaenoic acid) fatty acids are important for neural structure and functionReference Neuringer, Anderson and Conner95Reference Conklin, Gianaros and Brown101 and this aspect may explain studies where improvements in EDSS have also been reported. Furthermore requirements for essential polyunsaturated fatty acids increase as a function of the amount of saturated fat in the dietReference Holman102 and we have recently found significant positive correlations between dietary total saturated and total monounsaturated fatty acids and delta-6 and delta-5 desaturase gene expression in human PBMCReference Xiang, Rahman, Ai, Li and Harbige103. The level of dietary saturated and monounsaturated fatty acids should not therefore be ignored and may be important factors in some of the trials discussed above and relate to the MS epidemiological correlations, mentioned earlier in this overview, in relation to an increased requirement for polyunsaturated fatty acids in MS.

Conclusions and perspectives

Taken overall the epidemiological, biochemical, experimental animal model and clinical trial data described in this overview show that polyunsaturated fatty acids, particularly the n-6 fatty acids, do have a role in the pathogenesis and treatment of multiple sclerosis. We have demonstrated dysregulation of n-6 fatty acid metabolism and cytokines in MS and have been able to show in a small “proof of concept” clinical trial a marked therapeutic benefit. Thus we suggest that dysregulation of n-6 fatty acid metabolism and cytokines is one mechanism that is important in disease progression, which is modifiable by specific supplementation. Thus metabolic disturbance of the production of the long chain n-6 fatty acids DGLA and AA affects the physiological integrity of immune cells, in that they have a limited ability to produce TGF-β, under relapse conditions, which is important for the regulation of pro-inflammatory cytokine production e.g. TNF-α, IL-1β, IFN-γ as well as other cellular biological functions. It is also known that TGF-β and fatty acids such as arachidonic acid are important in the growth and differentiation of oligodendrocytes and in myelinationReference Sinclair and Crawford104Reference Van Meeteren, Baron, Beermann, Dijkstra and van Tol108 which would therefore be of importance in the stimulation, growth and recovery of these cells in MS. These findings provide a link between, dietary, metabolic, immunological and neurobiological aspects of MS and therefore for the first time we can begin to make sense of the wealth of apparently unconnected aspects of MS, particularly in relation to dietary fats. More basic research is still required such as characterisation of possible desaturase gene polymorphisms, lymphocyte desaturase gene expression and analysis of specific lymphocyte phospholipid classes and their fatty acid composition in relation to cytokine and chemokine gene expression and production. This should be undertaken in well defined MS patient groups e.g. active MS and primary and secondary chronic progressive forms of the disease and over an extended period of time. Furthermore large well controlled clinical trials with different doses of well characterised and safe fatty acid formulations as well as manipulation of dietary saturated fatty acids could be undertaken. Clinical trials should include MRI, MR spectroscopy and analysis of lesion burden and cortical gray matter density in order to investigate any possible effects of polyunsaturated fatty acids on myelination, neuronal, dentritic, glial and neurite packing densities. In addition biochemical monitoring of peripheral blood cell membrane phospholipid fatty acids, particularly lymphocytes, should be undertaken as well as immunological studies such as T-cell and macrophage pro- and anti-inflammatory cytokine gene expression and production, T regulatory cells and anti-myelin antibodies. In this way a more complete picture will emerge of the clinical and therapeutic significance and the metabolic, immunological and neurological bases to the role of polyunsaturated fatty acids in the pathogenesis and treatment of MS.

Conflict of interest statement

BGC20-884 and related intellectual property are patented by BTG International Ltd with LSH and MKS as named inventors. LH and MKS co-wrote the manuscript. At the time of the trial there were no conflicts of interests. Subsequently to the trial findings BGC20-884 and related intellectual property is now the subject of patents held by BTG International Ltd with LSH and MKS as named inventors. LH wrote the text and MKS was the lead trial neurologist.

References

1Ewing, C & Bernard, CC (1998) Insights into the aetiology and pathogenesis of multiple sclerosis. Immunol Cell Biol. 76, 4754.CrossRefGoogle ScholarPubMed
2Noseworthy, JH (1999) Progress in determining the causes and treatment of multiple sclerosis. Nature 399, Suppl. 24, A40A46.CrossRefGoogle ScholarPubMed
3Martino, G & Hartung, H-P (1999) Immunopathogenesis of multiple sclerosis: the role of T cells. Curr Opin Neurol 12, 309321.CrossRefGoogle ScholarPubMed
4Hafler, DA (2004) Multiple Sclerosis. J. Clin Invest. 113, 788794.CrossRefGoogle ScholarPubMed
5Fredrikson, S, Soderstrom, M, Hillert, J, et al. (1994) Multiple sclerosis: occurrence of myelin basic protein peptide-reactive T cells in healthy family members. Acta Neurol Scand 89, 184189.CrossRefGoogle ScholarPubMed
6Kerlero de Rosbo, N, Milo, R, Lees, MB, et al. (1993) Reactivity to myelin antigens in multiple sclerosis. Peripheral blood lymphocytes respond predominantly to myelin oligodendrocyte glycoprotein. J Clin Invest 92, 2602–8.CrossRefGoogle ScholarPubMed
7Kerlero de Rosbo, N, Hoffman, M, Mendel, I, et al. (1997) Predominance of the autoimmune response to myelin oligodendrocyte glycoprotein (MOG) in multiple sclerosis: reactivity to the extracellular domain of MOG is directed against three main regions. Eur J Immunol 27, 3059–69.CrossRefGoogle Scholar
8Chou, YK, Bourdette, DN, Offner, H, et al. (1992) Frequency of T cells specific for myelin basic protein and myelin proteolipid protein in blood and cerebrospinal fluid in multiple sclerosis. J Neuroimmunol 38, 105114.CrossRefGoogle ScholarPubMed
9Ota, K, Matsui, M, Milford, EL, et al. (1990) T cell recognition of an immunodominant myelin basic epitope in multiple sclerosis. Nature 346, 183187.CrossRefGoogle ScholarPubMed
10Burns, J, Bartholomew, B & Lobo, S (1999) Isolation of myelin basic protein-specific T cells predominantly from the memory T-cell compartment in multiple sclerosis. Ann Neurol 45, 3339.3.0.CO;2-G>CrossRefGoogle ScholarPubMed
11Zhang, J, Markovic-Plese, S, Lacet, B, et al. (1994) Increased frequency of interleukin 2-responsive T cells specific for myelin basic protein and proteolipid protein in peripheral blood and cerebrospinal fluid of patients with multiple sclerosis. J Exp Med 179, 973984.CrossRefGoogle ScholarPubMed
12Tejada-Simon, MV, Hong, J, Rivera, VM, et al. (2001) Reactivity pattern and cytokine profile of T cells primed by myelin peptides in multiple sclerosis and healthy individuals. Eur J Immunol 31, 907917.3.0.CO;2-1>CrossRefGoogle ScholarPubMed
13Bielekova, B, Goodwin, B, Richert, N, Cortese, I, Kondo, T, Afshar, G, Gran, B, Eaton, J, et al. (2000) Encephalitogenic potential of the myelin basic protein peptide (amino acids 83-99) in multiple sclerosis: results of a phase II clinical trial with an altered peptide ligand. Nature Medicine 6, 11671175.CrossRefGoogle ScholarPubMed
14Navikas, V & Link, H (1996) Review: cytokines and the pathogenesis of multiple sclerosis. J Neurosci Res 45, 322333.3.0.CO;2-B>CrossRefGoogle ScholarPubMed
15McCarron, RM, Wang, L, Racke, MK, et al. (1993) Cytokine-regulated adhesion between encephalitogenic T lymphocytes and cerebrovascular endothelial cells. J Neuroimmunol 43, 2330.CrossRefGoogle ScholarPubMed
16Selmaj, K, Raine, CS, Farooq, M, et al. (1991) Cytokine cytotoxicity against oligodendrocytes. Apoptosis induced by lymphotoxin. J Immunol 147, 15221529.CrossRefGoogle ScholarPubMed
17Vartanian, T, Li, Y, Zhao, M, et al. (1995) Interferon-gamma-induced oligodendrocyte cell death: implications for the pathogenesis of multiple sclerosis. Mol Med 1, 732743.CrossRefGoogle ScholarPubMed
18Kuroda, Y & Shimamoto, Y (1991) Human tumor necrosis factor-alpha augments experimental allergic encephalomyelitis in rats. J Neuroimmunol 34, 159164.CrossRefGoogle ScholarPubMed
19Issazadeh, S, Lorentzen, JC, Mustafa, MI, et al. (1996) Cytokines in relapsing experimental autoimmune encephalomyelitis in DA rats: persistent mRNA expression of proinflammatory cytokines and absent expression of interleukin-10 and transforming growth factor-beta. J Neuroimmunol 69, 103115.CrossRefGoogle ScholarPubMed
20Ruddle, NH, Bergman, CM, McGrath, KM, et al. (1990) An antibody to lymphotoxin and tumor necrosis factor prevents transfer of experimental allergic encephalomyelitis. J Exp Med 172, 11931200.CrossRefGoogle ScholarPubMed
21Cannella, B & Raine, CS (1995) The adhesion molecule and cytokine profile of multiple sclerosis lesions. Ann Neurol 37, 424435.CrossRefGoogle ScholarPubMed
22Merrill, JE, Strom, SR, Ellison, GW, et al. (1989) In vitro study of mediators of inflammation in multiple sclerosis. J Clin Immunol 9, 8496.CrossRefGoogle ScholarPubMed
23Maimone, D, Reder, AT & Gregory, S (1993) T cell lymphokine-induced secretion of cytokines by monocytes from patients with multiple sclerosis. Cell Immunol 146, 96106.CrossRefGoogle ScholarPubMed
24Hirsch, RL, Panitch, HS & Johnson, KP (1985) Lymphocytes from multiple sclerosis patients produce elevated levels of gamma interferon in vitro. J Clin Immunol 5, 386389.CrossRefGoogle ScholarPubMed
25Hollifield, RD, Harbige, LS, Pham-Dinh, D & Sharief, M (2003) Evidence for cytokine dysregulation in multiple sclerosis: peripheral blood mononuclear cell production of pro-inflammatory and anti-inflammatory cytokines during relapse and remission. Autoimmunity 36, 133141.CrossRefGoogle ScholarPubMed
26Imamura, K, Suzumura, A, Hayashi, F, et al. (1993) Cytokine production by peripheral blood monocytes/macrophages in multiple sclerosis patients. Acta Neurol Scand 87, 281285.CrossRefGoogle ScholarPubMed
27Philippe, J, Debruyne, J, Leroux-Roels, G, et al. (1996) In vitro TNF-alpha, IL-2 and IFN-gamma production as markers of relapses in multiple sclerosis. Clin Neurol Neurosurg 98, 286290.CrossRefGoogle ScholarPubMed
28Bertolotto, A, Malucchi, S, Capobianco, M, et al. (1999) Quantitative PCR reveals increased levels of tumor necrosis factor-alpha mRNA in peripheral blood mononuclear cells of multiple sclerosis patients during relapses. J Interferon Cytokine Res 19, 575581.CrossRefGoogle ScholarPubMed
29Beck, J, Rondot, P, Catinot, L, et al. (1988) Increased production of interferon gamma and tumor necrosis factor precedes clinical manifestation in multiple sclerosis: do cytokines trigger off exacerbations? Acta Neurol Scand 78, 318323.CrossRefGoogle ScholarPubMed
30Sharief, MK & Hentges, R (1991) Association between tumour necrosis factor-alpha and disease progression in patients with multiple sclerosis. N. Engl. J. Med. 325, 467472.CrossRefGoogle ScholarPubMed
31Sharief, MK & Thompson, EJ (1992) In vivo relationship of tumor necrosis factor-alpha to blood-brain barrier damage in patients with active multiple sclerosis. J Neuroimmunol 38, 2733.CrossRefGoogle ScholarPubMed
32Rieckmann, P, Albrecht, M, Kitze, B, et al. (1994) Cytokine mRNA levels in mononuclear blood cells from patients with multiple sclerosis. Neurology 44, 15231526.CrossRefGoogle ScholarPubMed
33Lu, CZ, Jensen, MA & Arnason, BG (1993) Interferon gamma- and interleukin-4-secreting cells in multiple sclerosis. J Neuroimmunol 46, 123128.CrossRefGoogle ScholarPubMed
34Bertolotto, A, Capobianco, M, Malucchi, S, et al. (1999) Transforming growth factor beta1 (TGFbeta1) mRNA level correlates with magnetic resonance imaging disease activity in multiple sclerosis patients. Neurosci Lett 263, 21–4.CrossRefGoogle ScholarPubMed
35Mokhtarian, F, Shi, Y, Shirazian, D, et al. (1994) Defective production of anti-inflammatory cytokine, TGF-beta by T cell lines of patients with active multiple sclerosis. J Immunol 152, 60036010.CrossRefGoogle Scholar
36De Stefano, N, Narayanan, S, Francis, GS, Arnaoutelis, R, Tartaglia, MC, Antel, JP, Matthews, PM & Arnold, DL (2001) Evidence of axonal damage in the early stages of multiple sclerosis and its relevance to disability. Arch Neurol. 58, 6570.CrossRefGoogle ScholarPubMed
37Bjartmar, C, Wujek, JR & Trapp, BD (2003) Axonal loss in the pathology of MS: consequences for understanding the progressive phase of the disease. J. Neuro. Sci. 206, 165171.CrossRefGoogle ScholarPubMed
38Lassmann, H, Bruck, W & Lucchinettii, C (2001) Heterogeneity of multiple sclerosis pathogenesis: implications for diagnosis and therapy. Trends in Molecular Medicine. 7, 115121.CrossRefGoogle ScholarPubMed
39Matute, C & Perez-Cerda, F (2005) Multiple sclerosis: novel perspectives on newly forming lesions. Trends in Neurosciences 28, 173175.CrossRefGoogle ScholarPubMed
40Swank, RL (1950) Multiple sclerosis: a correlation of its incidence with dietary fat. Am. J. Med. Sci. 220, 421430.CrossRefGoogle ScholarPubMed
41Alter, M, Yamoor, M & Harshe, M (1974) Multiple sclerosis and nutrition. Arch. Neurol. 31, 267272.CrossRefGoogle ScholarPubMed
42Wolfgram, F (1975) Similar geographical distribution of multiple sclerosis and cancer of the colon. Acta. Neurol. Scandinav. 52, 294302.CrossRefGoogle ScholarPubMed
43Agranoff, BW & Goldberg, D (1974) Diet and the geographical distribution of multiple sclerosis. Lancet, Nov 210611066.CrossRefGoogle ScholarPubMed
44Ghadirian, P, Jain, M, Ducic, S, et al. (1998) Nutritional factors in the aetiology of multiple sclerosis: a case-control study in Montreal, Canada. Int J Epidemiol. 27, 845852.CrossRefGoogle ScholarPubMed
45Murrell, TGC, Harbige, LS & Robinson, IC (1991) A review of the aetiology of multiple sclerosis: an ecological approach. Ann Hum Biol. 18, 95112.CrossRefGoogle ScholarPubMed
46Malosse, D, Perron, H, Sasco, A & Seigneurin, JM (1992) Correlation between milk and dairy product consumption and multiple sclerosis prevalence: a worldwide study. Neuroepidemiology 11, 304312.CrossRefGoogle ScholarPubMed
47Esparza, ML, Sasaki, S & Kesteloot, H (1995) Nutrition, latitude, and multiple sclerosis mortality: an ecologic study. Am. J. Epidemiol. 142, 733737.Google ScholarPubMed
48Zhang, SM, Willet, WC, Hernan, MA, Olek, MJ & Ascherio, A (2000) Dietary fat in relation to risk of multiple sclerosis among two large cohorts of women. Am J Epidemiol 152, 10561064.CrossRefGoogle ScholarPubMed
49Baker, RWR, Thompson, RHS & Zilkha, KJ (1964) Serum fatty acids in multiple sclerosis. J Neuro Neurosurg Psychiatry 27, 408414.CrossRefGoogle ScholarPubMed
50Sanders, H, Thompson, RH, Wright, HP & Zilkha, KJ (1968) Further studies on platelet adhesiveness and serum cholesteryl linoleate levels in multiple sclerosis. J. Neurol. Neurosurg. Psychiat. 31, 321325.CrossRefGoogle ScholarPubMed
51Gul, S, Smith, AD, Thompson, RHS, et al. (1970) Fatty acid composition of phospholipids from platelets and erythrocytes in multiple sclerosis. J Neurol Neurosurg Psychiat 33, 506510.CrossRefGoogle ScholarPubMed
52Thompson, RHS (1973) Fatty acid metabolism in multiple sclerosis. Biochemical Society Symposium 35, 103111.Google Scholar
53Thompson, RHS (1975) Unsaturated fatty acids in multiple sclerosis. In Multiple Sclerosis Resaerch, pp. 184193 [Davison, AN, Humphrey, JH, Liversedge, AL, McDonald, WI and Porterfield, JS, editors]. USA, Elsevier: North Holland.Google Scholar
54Tsang, WM, Belin, J, Monro, JA, Smith, AD, Thompson, RHS & Zilkha, KJ (1976) Relationship between plasma and lymphocyte linoleate in multiple sclerosis. J Neurol Neurosurg Psychiatry 39, 767771.CrossRefGoogle ScholarPubMed
55Homa, ST, Belin, J, Smith, AD, et al. (1980) Levels of linoleate and arachidonate in red blood cells of healthy individuals and patients with multiple sclerosis. J Neurol Neurosurg Psychiat 43, 106110.CrossRefGoogle ScholarPubMed
56Neu, IS (1983) Essential fatty acids in the serum and cerebrospinal fluid of multiple sclerosis patients. Acta. Neurol. Scand. 67, 151163.CrossRefGoogle ScholarPubMed
57Cherayil, GD (1984) Sialic acid and fatty acid concentrations in lymphocytes, red blood cells, and plasma from patients with multiple sclerosis. J Neuro Sci. 63, 110.CrossRefGoogle ScholarPubMed
58Fisher, M, Johnson, MH, Natale, AM, et al. (1987) Linoleic acid levels in white blood cells, platelets and serum of multiple sclerosis patients. Acta Neurol Scand 76, 241245.CrossRefGoogle ScholarPubMed
59Navarro, X & Segura, R (1989) Red blood cell fatty acids in multiple sclerosis. Acta Neurol Scand 79, 3237.CrossRefGoogle ScholarPubMed
60Holman, RT, Johnson, SB & Kokmen, E (1989) Deficiencies of polyunsaturated fatty acids and replacement by nonessential fatty acids in plasma lipids in multiple sclerosis. Proc. Natl. Acad. Sci. USA. 86, 47204724.CrossRefGoogle ScholarPubMed
61Shukla, VKS & Clausen, J (1978) Linoleate and fatty acid patterns of serum lipids in multiple sclerosis. Acta. Neurol. Scandinav 57, 270274.CrossRefGoogle ScholarPubMed
62Yoshida, M, Takase, S, Itahara, K & Nakanishi, T (1983) Linoleate and fatty acid compositions in the serum lipids of Japanese patients with multiple sclerosis. Acta Neurol Scand 68, 362364.CrossRefGoogle ScholarPubMed
63Heipertz, R, Klauke, W, Pilz, H & Ritter, G (1977) Serum fatty acids in multiple sclerosis. J. Neurology 214, 153157.CrossRefGoogle ScholarPubMed
64Nightingale, S, Woo, E, Smith, AD, et al. (1990) Red blood cell and adipose tissue fatty acids in mild inactive multiple sclerosis. Acta Neurol Scand 82, 4350.CrossRefGoogle ScholarPubMed
65Love, WC, Cashell, A, Reynolds, M & Callaghan, N (1974) Linoleate and fatty acid patterns of serum lipids in multiple sclerosis and other diseases. Br Med J 3, 1821.CrossRefGoogle ScholarPubMed
66Jones, R & Harbige, LS (1987) Erythrocytes in multiple sclerosis: effect of increased intake of essential fatty acids on phosphoglycerides and electrophoretic mobility. In Multiple Sclerosis, Immunological, Diagnostic and Therapeutic Aspects, pp. 201209 [Clifford Rose, F and Jones, R, editors]. London: John Libbey & Co Ltd.Google Scholar
67Harbige, LS, Crawford, MA, Jones, R, Preece, AW & Forti, A (1986) Dietary intervention studies on the phosphoglyceride fatty acids and electrophoretic mobility of erythrocytes in multiple sclerosis. Prog. Lipid Res 25, 243248.CrossRefGoogle Scholar
68Homa, ST, Conroy, DM, Belin, J, Smith, AD, Monro, JA & Zilkha, KJ (1981) Fatty acid patterns of red blood cell phospholipids in patients with multiple sclerosis. Lancet, August 29474.CrossRefGoogle ScholarPubMed
69Nordvik, I, Myhr, K-M, Nyland, H & Bjerve, KS (2000) Effect of dietary advice and n-3 supplementation in newly diagnosed MS patients. Acta. Neurol Scand 102, 143–149.CrossRefGoogle ScholarPubMed
70Field, EJ & Joyce, G (1983) Multiple sclerosis: effect of gamma-linolenate administration upon membranes and the need for extended clinical trials of unsaturated fatty acids. Eur. Neurol. 22, 78–83.CrossRefGoogle ScholarPubMed
71Field, EJ, Joyce, G & Smith, BM (1977) Erythrocyte-UFA (Eufa) mobility test for pathogenesis and handling of the disease. J Neurol 214, 113127.CrossRefGoogle ScholarPubMed
72Harbige, LS (2003) Fatty acids, the immune response, and autoimmunity: a question of n-6 essentiality and the balance between n-6 and n-3. Lipids 38, 323341.CrossRefGoogle ScholarPubMed
73Meade, CJ, Mertin, J, Sheena, J & Hunt, R (1978) Reduction by linoleic acid of the severity of experimental allergic encephalomyelitis in the guinea-pig. J Neuro Sci 35, 291–308.CrossRefGoogle ScholarPubMed
74Mertin, J & Stackpoole, A (1978) Suppression by essential fatty acids of experimental allergic encephalomyelitis is abolished by indomethacin. Prostaglandins and Medicine 1, 283291.CrossRefGoogle ScholarPubMed
75Harbige, LS, Yeatman, N, Amor, S & Crawford, MA (1995) Prevention of experimental autoimmune encephalomyelitis in Lewis rats by a novel source of γ-linolenic acid. Br J Nutr 74, 701715.CrossRefGoogle ScholarPubMed
76Harbige, LS, Layward, L, Morris-Downes, MM, et al. (2000) The protective effects of omega-6 fatty acids in experimental autoimmune encephalomyelitis (EAE) in relation to transforming growth factor-beta 1 (TGF-beta1) up-regulation and increased prostaglandin E2 (PGE2) production. Clin Exp Immunol 122, 445–452.CrossRefGoogle ScholarPubMed
77Adda, DH, Beraud, E & Depieds, R (1977) Evidence for suppressor cells in Lewis rats' experimental allergic encephalomyelitis. Eur J Immunol 7, 620623.CrossRefGoogle ScholarPubMed
78Karpus, WJ & Swanborg, RH (1991) CD4+ suppressor cells inhibit the function of effector cells of experimental autoimmune encephalomyelitis through a mechanism involving transforming growth factor-β. J Immunol 146, 11631168.CrossRefGoogle ScholarPubMed
79Rack, MK, Sriram, S, Calrlini, J, Cannella, B, Raine, CS & McFarlin, DE (1993) Long-term treatment of chronic relapsing experimental allergic encephalomyelitis by transforming growth factor-β2. J Neuroimmunol 46, 175–183.CrossRefGoogle Scholar
80Santambrogio, L, Hochwald, GM, Saxena, B, Leu, CH, Martz, JE, Carlino, JA, Ruddle, NH, Palladino, MA, Gold, LI & Thorbecke, GJ (1993) Studies on the mechanisms by which Transforming Growth Factor-β protects against allergic encephalomyelitis. J Immunol 151, 11161127.CrossRefGoogle ScholarPubMed
81Ovadia, H & Paterson, PY (1982) Effect of indomethacin treatment upon actively-induced and transferred experimental allergic encephalomyelitis (EAE) in Lewis rats. Clin Exp Immunol 49, 386–392.Google ScholarPubMed
82Liblau, RS, Singer, SM & McDevitt, (1995) Th1 and Th2 CD4+T cells in the pathogenesis of organ-specific autoimmune diseases. Immunology Today 16, 34–38.CrossRefGoogle ScholarPubMed
83Khoury, SJ, Hancock, WW & Weiner, HL (1992) Oral tolerance to myelin basic protein and natural recovery from experimental autoimmune encephalomyelitis are associated with downregulation of inflammatory cytokines and differential upregulation of transforming growth factor β, interleukin 4, and prostaglandin E expression in the brain. J Exp Med 176, 13551364.CrossRefGoogle ScholarPubMed
84Bates, D, Fawcett, PRW, Shaw, DA & Weightman, D (1978) Polyunsaturated fatty acids in the treatment of acute remitting multiple sclerosis. Br Med J. 2, 13901391.CrossRefGoogle ScholarPubMed
85Millar, JHD, Zilkha, KJ, Langman, MJS, Payling-Wright, H, et al. (1973) Double-blind trial of linoleate supplementation of the diet in multiple sclerosis. Br Med J 1, 765–768.CrossRefGoogle ScholarPubMed
86Paty, DW, Cousin, HK, Read, S & Adlakha, K (1978) Linoleic acid in multiple sclerosis: failure to show any therapeutic benefit. Acta Neuro Scand 58, 53–58.CrossRefGoogle ScholarPubMed
87Dworkin, RH, Bates, D, Millar, JHD, et al. (1984) Linoleic acid and multiple sclerosis: a reanalysis of three double blind trials. Neurology 34, 14411445.CrossRefGoogle ScholarPubMed
88Bates, D, Cartlidge, NEF, French, JM, et al. (1989) A double-blind controlled trial of long chain n-3 polyunsaturated fatty acids in the treatment of multiple sclerosis. J Neurol Neurosurg Psychiatry 52, 18–22.CrossRefGoogle ScholarPubMed
89Swank, RL (1970) Multiple sclerosis: twenty years on a low fat diet. Arch. Neurol. 23, 460474.CrossRefGoogle ScholarPubMed
90Fitzgerald, G, Harbige, LS, Forti, A & Crawford, MA (1987) The effect of nutritional counselling on diet and plasma EFA status in multiple sclerosis patients over 3 years. Numan Nutrition: Applied Nutrition, 41A, 297–310.Google ScholarPubMed
91Swank, RL & Grimsgaard, A (1988) Multiple sclerosis: the lipid relationship. Am. J. Clin. Nutr 48, 13871393.CrossRefGoogle ScholarPubMed
92Swank, RL & Dugan, BB (1990) Effect of low saturated fat diet in early and late cases of multiple sclerosis. Lancet, July 7 336, 37–39.CrossRefGoogle ScholarPubMed
93Harbige, LS, Jones, R, Jenkins, R, Fitzgerald, G, Forti, A & Budowski, P (1990) Nutritional management in multiple sclerosis with reference to experimental models. Ups J Med Sci 48, 189207.Google ScholarPubMed
94Harbige, LS, Hollifield, RD, Pinto, E, Xiang, M, Leach, M & Sharief, MK (2007) Polyunsaturated fatty acids (n-6) in the treatment and pathogenesis of multiple sclerosis: ii results of a randomised, double blind, placebo controlled trial for. Lancet.Google Scholar
95Neuringer, M, Anderson, GJ & Conner, WE (1988) The essentiality of n-3 fatty acids for the development and function of the retina and brain. Ann. Rev. Nutr. 8, 517541.CrossRefGoogle ScholarPubMed
96Birch, EE, Garfield, S, Castaneda, Y, Hughbanks-Wheaton, D, Uauy, R & Hoffman, D (2007) Visual acuity and cognitive outcomes at 4 years of age in a double-blind, randomized trial of long-chain polyunsaturated fatty acid-supplemented infant formula. Early. Hum. Dev. 83, 279–284.CrossRefGoogle Scholar
97Martinez, M, Vazquez, E, Garcia-Silva, MT, et al. (2000) Therapeutic effects of docosahexaenoic acid ethyl ester in patients with generalized peroxisomal disorders. Am. J. Clin. Nutr 71, Suppl, 376S–85S.CrossRefGoogle ScholarPubMed
98Crawford, MA, Costeloe, K, Ghebremeskel, K, et al. (1997) Are deficits of arachidonic and docosahexaenoic acids responsible for the neural and vascular complications of preterm babies. Am. J. Clin. Nutr. 66, Suppl, 1032S–41S.CrossRefGoogle ScholarPubMed
99Xiang, M, Alfven, G, Blennow, M, Trygg, M & Zetterstrom, R (2000) Long chain polyunsaturated fatty acids in human milk and brain growth during early infancy. Acta Paediatr 89, 142–147.CrossRefGoogle ScholarPubMed
100De la Pressa Owens, S & Innis, SM (2000) Diverse, region specific effects of addition of arachidonic and docosahexaenoic acids to formula with low or adequate linoleic and alpha-linolenic acids on piglet brain monoaminergic neurotransmitters. Pediatr Res 48, 125–130.CrossRefGoogle Scholar
101Conklin, SM, Gianaros, PJ, Brown, SM, et al. (2007) Long-chain omega-3 fatty acid intake is associated positively with corticolimbic grey matter volume in healthy adults. Neurosci Lett 421, 209212.CrossRefGoogle ScholarPubMed
102Holman, RT (1960) The ratio of trienoic: tetraenoic acids in tissue lipids as a measure of essential fatty acid requirements. J. Nutr 70, 405410.CrossRefGoogle Scholar
103Xiang, M, Rahman, MA, Ai, H, Li, X & Harbige, LS (2006) Diet and gene expression: delta-5 and delta-6 desaturases in healthy Chinese and European subjects. Ann Nutr Metab 50, 492498.CrossRefGoogle ScholarPubMed
104Sinclair, AJ & Crawford, MA (1972) The accumulation of arachidonate and docosahexaenote in the developing rat brain. J. Neurochem 19, 17531758.CrossRefGoogle ScholarPubMed
105Merrill, JE & Zimmerman, RP (1991) Natural and induced cytotoxicity of oligodendrocytes by microglia is inhibitable by TGF beta. Glia 4, 327–331.CrossRefGoogle ScholarPubMed
106Copeland, C, Curzner, ML, Groome, N & Diemel, LT (2000) Temporal analysis of growth factor mRNA expression in myelinating rat brain aggregate cultures: increments in CNTF, FGF-2, IGF-I, and PDGF-AA mRNA are induced by antibody-mediated demyelination. Glia 30, 342351.Google Scholar
107Serafina, S, Sanchez, M, Campeggi, L, Suchanek, G, Breitschop, H & Lassmann, H (1996) Accelerated myelinogenesis by dietary lipids in rat brain. Journal Neurochemistry 67, 17441750.Google Scholar
108Van Meeteren, ME, Baron, W, Beermann, C, Dijkstra, CD & van Tol, EA (2006) Polyunsaturated fatty acid supplementation stimulates differentiation of oligodendroglia cells. Dev Neurosci 28, 196–208.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 Geographical distribution of multiple sclerosis (from Adams C (1989) colour atlas of multiple sclerosis and other myelin disorders, wolfe medical publications ltd, with permission).

Figure 1

Fig. 2 Relationship between linoleic acid (18 : 2n-6) and arachidonic acid (20 : 4n-6) in peripheral blood mononuclear cell total phospholipids of healthy controls and multiple sclerosis.

Figure 2

Fig. 3 Relationship between linoleic acid (18 : 2n-6) and dihomo-γ-linolenic acid (20 : 3n-6) in peripheral blood mononuclear cell total phospholipids of healthy controls and multiple sclerosis.

Figure 3

Fig. 4 Relationship between dihomo-γ-linolenic acid (20 : 3n-6) and arachidonic acid (20 : 4n-6) in peripheral blood mononuclear cell total phospholipids of healthy controls and multiple sclerosis.

Figure 4

Fig. 5 Mean annualised relapse rate per patient for multiple sclerosis patients receiving high (n = 11) and low dose (n = 7) GLA-rich oil or placebo control (n = 10) over 18 months.

Figure 5

Fig. 6 Disability progression as measured by the EDSS (Expanded Disability Status Scale) in multiple sclerosis patients receiving high (n = 11) and low dose (n = 7) GLA-rich oil or placebo control (n = 10) over 18 months.