Skip to main content
Lippincott Open Access logoLink to Lippincott Open Access
. 2023 Apr 27;5(2):e00022. doi: 10.1097/IN9.0000000000000022

Overview of diet and autoimmune demyelinating optic neuritis: a narrative review

Scott M Plafker 1,2,*, Tyler Titcomb 3, Katarzyna Zyla-Jackson 1,2, Aneta Kolakowska 1, Terry Wahls 3
PMCID: PMC10144304  PMID: 37128292

Abstract

This review summarizes the cellular and molecular underpinnings of autoimmune demyelinating optic neuritis (ADON), a common sequela of multiple sclerosis and other demyelinating diseases. We further present nutritional interventions tested for people with multiple sclerosis focusing on strategies that have shown efficacy or associations with disease course and clinical outcomes. We then close by discuss the potential dietary guidance for preventing and/or ameliorating ADON.

Keywords: multiple sclerosis, diet, nutrition, optic neuritis, optic nerve, experimental autoimmune encephalomyelitis

1. Introduction

Autoimmune diseases have been increasing [1,2], with 5% to 8% of the US population having one or more of these disorders [3]. This rapid upward trajectory implicates improved diagnostics and changes in environmental risk factors such as diet and lifestyle. The deleterious impacts of diet have in part been attributed to the excessive consumption of ultra-processed foods enriched in simple carbohydrates and pro-inflammatory fats. Consuming ultra-processed foods can cause vitamin and mineral deficiencies [4,5], disrupt microbiome–host symbiosis, promote systemic inflammation, and drive insulin resistance and metabolic syndrome [68], all of which exacerbate autoimmune sequelae. Moreover, tissue-specific and systemic immune responses are highly integrated with metabolic and gut microbiome status (eg, [914]) as the mammalian gastrointestinal tract hosts the largest reservoir of immune cells in the body [15].

Here, we review the impacts of dietary approaches and components on non-ocular multiple sclerosis (MS) sequelae and consider their applications for MS-autoimmune demyelinating optic neuritis (MS-ADON) based on the candidate mechanisms underlying autoimmune-mediated optic nerve (ON) damage. Ocular pathologies are relatively understudied sequelae of autoimmunity with ADON being the most prevalent visual complication in people with multiple sclerosis (pwMS) [16], neuromyelitis optica spectrum disorder (NMOSD) [17], and myelin oligodendrocyte glycoprotein antibody-associated diseases (MOG-AD) [1820]. ADON affects ~50% of pwMS and is the first demyelinating event (FDE) for ~20% [2123]. The prevalence of ADON at disease onset for NMOSD and MOG-AD is 50% and 74%, respectively [17,24,25]. Because the most information regarding dietary interventions comes from the MS literature, this review focuses on MS-associated ADON and the potential of nutritional interventions.

ADON results from inflammation and demyelination of the ON, disrupting impulse conduction from the nerve to the visual cortex. Clinical hallmarks include pain, blurred vision, color desaturation, and loss of visual acuity and contrast sensitivity. Axons comprising the ON extend from retinal ganglion cells (RGCs), and demyelination of these axons induces RGC apoptosis. ADON is commonly monocular and lesion localization is retrobulbar in MS [26]. Although most pwMS recover vision following the resolution of inflammation, recurrent flare-ups can culminate in irreversible vision loss. First-line treatment for acute attacks is intravenous steroids, which accelerate recovery for some patients but fail to impact relapse frequency [27]. Second-line treatments include plasma exchange, IV immune globulin, and other agents (reviewed in [17,26,2830]). Proper diagnosis is paramount as several highly-prescribed MS treatments (eg, glatiramer acetate, interferon β (IFN-β), fingolimod, and dimethyl fumarate) can worsen conditions for people with NMO or MOG-ADs [17,31,32].

2. Cellular and molecular mechanisms of MS-ADON

A brief synopsis follows of the immune cell types, cytokines, and chemokines most associated with MS-ADON, based on the biofluid and immunohistological findings from pwMS and pre-clinical studies. Notably, the pre-clinical studies derive largely from experimental autoimmune encephalomyelitis (EAE), a widely used rodent model for human autoimmune demyelinating diseases of the CNS. Inducible versions of EAE use antigens derived from MOG, myelin basic protein, or proteolipid protein with symptoms modeling numerous motor and visual pathologies of human MS [33]. Transgenic models include (a) the 2D2 mouse that expresses a T-cell receptor for MOG peptide 35–55 and results in age-dependent, isolated optic neuritis [34]; and (b) the OSE opticospinal double strain modeling NMOSD [35].

2.1 IL-17-secreting T cells

IL-17A/F-secreting cells maintain gut homeostasis and communicate gut health throughout the body [36] but can transition to pathogenicity and promote CNS pathologies in MS and EAE [37,38]. pwMS have increased IL-17 mRNA in their blood and cerebrospinal fluid during relapses [39]. This cytokine is produced by effector T cell types including TH17, TH1, γδ-T cells, ex-Tregs, CD8+ Tc17 cells, natural killer T cells, and group 3 innate lymphoid cells [4043]. In the context of ADON, adoptively-transferred myelin-specific TH17 cells cause more severe optic neuritis and visual deficits than the adoptively-transferred TH1 cells [44], although both cell types drive disease in MS and EAE [44,45]. In fact, IL-17 mRNA expression levels in EAE mice are highest in the ON, compared with the brain and spinal cord, and neutralizing antibodies against IL-17 ablate ADON-mediated structural damage to the ON but are less effective at reducing motor deficits [46]. Additionally, effector T cells on ONs are enriched in markers indicative of TH17 transcriptional programming (eg, IL1r and IL23r). γδ-T cells that secrete IL-17 and express the chemokine receptor CCR6 were approximately three-fold higher on the ONs of EAE mice, compared with the brain or spinal cord [46]. Additional pro-inflammatory roles for IL-17 in ADON include recruiting and activating Ly6G+ neutrophils to the ONs and brains of EAE mice via the subarachnoid space [46]. IL-17 also limits (re)myelination by suppressing the maturation of NG2+ oligodendrocyte precursors [47].

2.2 CD8+T cells

The contributions of CD8+ T cells to MS have been reviewed [45,48,49]. Highlights include the following: (a) select major histocompatibility complex (MHC) Class I human leukocyte antigen (HLA) alleles conferring increased or decreased genetic risk for disease (eg, [5053]); and (b) CD8+ T cells being enriched in the CSF [54] and at lesion edges [55] and often in greater abundance at sites of infiltrates than CD4+ T cells (eg, [56,57]). Contributions of CD8+ T cells to ADON have been reported for MS [58], but more so for NMOSD (eg, [5860]); however, reduced naïve CD8+ T cells and increased effector/memory CD8+ T cells in the circulation have been detected for both diseases [61].

2.3 Astrocytes

ON astrocytes play a central role in ADON in the EAE model (eg, [6264]) with activated astrocytes detectable as an early marker and inducer of inflammation, via NF-кB, on ONs, before frank immune cell infiltration [64]. Elevated lipocalin-2 in EAE studies and patient sera [63] along with increased complement cascade components, especially C3 and SerpinG1, and suppressed cholesterol biosynthesis machinery were identified as major changes in ON astrocytes contributing to pathogenicity [64]. Curiously, the change in C3 was specific to females and correlated with upregulation of the C3 receptor on microglia/macrophages. These EAE findings fit with C3 knockout mouse studies [65,66] and complement deposition being detected in MS brain and spinal cord lesions [67]. Reduced cholesterol biosynthesis may underlie compromised remyelination of the ON because astrocytes provide cholesterol to oligodendrocytes for myelin synthesis and to neurons for building synapses and membranes. Fitting with this, enhancing cholesterol transport using gentisic acid ameliorated ON damage and spared vision and ocular structure in EAE mice [68].

2.4 B cells

B cells can present antigen to activate T cells during the earliest stages of ADON, and select B cell-related markers including CD19, CD20, and CD79A in peripheral blood mononuclear cells have been associated with first episodes of ADON. Specifically, higher expression levels of these markers correlated with the severity of visual acuity deficits [69,70]. Interestingly, these markers largely did not differ between subjects irrespective of the extent of vision recovery [69]. In addition, naïve CD19+ CD24+ CD38+ B regulatory cells were higher in people experiencing ADON when compared with age-matched, healthy controls, although differences between the cohorts were not detected in the capacity of these cells to produce IL-10, a primary anti-inflammatory cytokine that suppresses pathogenic T cells through multiple mechanisms [71].

2.5 Macrophages and microglia

Macrophages and microglia are abundant in MS lesions (eg, [7275]) and blood [76] and are major drivers of inflammation and pathology early in EAE (eg, [72,7781]) and in the later stages of disease characterized by gliosis [82]. The involvement of these cell types in ON and retinal inflammation in EAE animals involves NF-кB [62], the secretion of IL-6 [83,84], TNF-α [85], inducible nitric oxide synthase [85], reactivation within the CNS of auto-reactive T cells, and suppression of tolerance-promoting Tregs (reviewed in [86]). Suppression of macrophage/microglia activation on ONs by inhibition of spermine oxidase [87] or genetic ablation of arginase 2 [88] in EAE mice improved visual acuity and reduced macrophage infiltration of ONs. Further, nicotinamide adenine dinucleotide supplementation mitigated microglial and astrocyte numbers on ONs and preserved myelination [89]. In contrast to these pro-inflammatory effector functions of M1-type macrophages, subsets of these cell types (ie, M2-type) are critical for resolving inflammation and promoting repair [90,91]. For example, activated resident microglia promote oligodendrocyte maturation and remyelination [92], and remove myelin and other cellular debris to assist with regeneration [81,93].

2.6 Infiltrating cell types in ADON lesions and CSF cytokines

The contributions of specific infiltrating cell types to MS-ADON lesions were recently analyzed through immunohistochemical-based immunoprofiling within different lesions (ie, active, chronic active, and inactive) in the parenchyma, meninges, and perivascular regions from post-mortem MS ONs. This study showed that (i) HLA-DR+ CD68+ myeloid cells were enriched in active lesions and along the edges of chronic active lesions; (ii) CD4+ T cells were elevated within chronic active lesions in the parenchyma and within all lesion types in the meninges; (iii) CD8+ T cells were detected in various lesions, outnumbered CD4+ T cells, but did not reach significance compared with controls; (iv) plasma cells were not enriched in most lesions irrespective of anatomical compartment, but CD138+ B cells were lower in active and chronic active parenchymal lesions whereas CD20+ B cells were elevated in active perivascular lesions; both markers were increased along the edges of chronic active perivascular lesions. In a study comparing cytokine profiles in the CSF from people experiencing ADON, levels of TNF-α, IL-10, and CXCL13 were higher in those individuals that progressed to a diagnosis of MS as compared to those with isolated optic neuritis that did not progress to MS [94]. The cohort that progressed to MS additionally had increased IgG indices and oligoclonal bands.

3. Dietary studies in animal models of MS

Mounting evidence demonstrates profound dietary impacts on EAE, although most studies have not assessed ocular sequelae [95110]. In those addressing ocular pathology, supplementation with a fatty acid (FA) cocktail of palmitic, oleic, stearic, linoleic, α-linolenic, γ-linolenic, eicosapentaenoic, and docosahexaenoic acids starting the day of MOG35-55-EAE immunization counteracted inflammatory and gliotic processes in the retina, reduced multiple inflammatory factors (TNF-α, IL-1β, IL-6, IL-8), and prevented RGC apoptosis [111]. This FA mixture restored the number of myelinated fibers and preserved photopic negative response amplitudes, a readout of inner retinal function. Neuroprotection was attributed to macrophage-derived oncomodulin, a trophic factor for neurons, and to a decreased ratio of pro-inflammatory (M1) to anti-inflammatory (M2) macrophages [112]. Individual FAs also repress disease as shown for conjugated linoleic acid in the OSE model [113] and valproic acid in EAE rats [114].

α-lipoic acid (α-LA) dramatically reduced MOG35-55-EAE-induced axonal injury in ONs, reduced CD4+ T lymphocytes and CD11b+ macrophages/microglia infiltrates, prevented inner retinal layer thinning, RGC apoptosis, and vision loss whether administered before or after symptom onset [115]. However, a second study showed that α-LA efficacy required administration either before or on the day of MOG immunization to preserve vision and did not prevent ON demyelination or reduce infiltrates [116]. We have shown that a weight-stabilizing ketogenic diet enriched in medium chain triglycerides, flaxseed oil, and insoluble fiber prevents the onset of optic neuritis and motor deficits in MOG35–55-EAE mice. When initiated after symptom onset, this diet improved visual and motor function in both male and female mice within 4 days and effectively resolved functional deficits tested within 2 weeks [117].

4. Human MS dietary studies

4.1 Dietary impacts on FDEs

The intersection of diet and MS has been garnering increased attention (Figure 1) but to date, no published human studies have specifically addressed the dietary impacts on ADON risk or resolution. However, studies have investigated the relationship between diet and FDEs, for which ≥20% of cases are expected to be ADON [2123]. Data from the multi-center, case-control, Ausimmune Study showed that higher yogurt, non-processed red meat, and fish consumption associated with decreased odds of FDE [118120]. Likewise, the Mediterranean diet and other healthy dietary patterns also associated with reduced odds of FDE in case-control studies [121,122]. In contrast, pro-inflammatory diets (based on the dietary inflammation index [123]) enriched in added sugars and ultra-processed foods are associated with increased FDE odds [124] and increased risk of autoimmune demyelinating diseases [125,126]. Corroborating these findings, a case-control study reported that low intake of fiber, vitamin D, and α-linolenic acid associated with increased odds of FDE [127].

Figure 1.

Figure 1.

PubMed search results for [Diet AND “multiple sclerosis”] conducted on January 3, 2023. Figure excludes incomplete numbers from 2023.

4.2 Foods associated with improved or worsened MS

The consumption of select foods has been correlated with MS disease trajectory. Higher fiber, fruits, vegetables, and healthy fat intake was positively associated with most health outcomes [128], and higher consumption of fruits, vegetables, beans, cherries, vitamin D, zinc, vitamin A, calcium, and vitamin B6 associated with reduced MS risk [129132]. Higher intake of full-fat dairy associated with lower T2 lesion volumes and higher intake of marine-derived omega-3 FAs aligned with greater normal appearing white matter microstructural integrity [133]. Similarly, a high prudent diet score, consisting of fruits, vegetables, fish, whole grains, and nuts, associated with reduced relapse risk in early MS in a 5-year longitudinal study [134]. A cross-sectional study among 2063 adults with MS revealed a link between higher adherence to Australian dietary guidelines for cardiovascular disease and lower relapse risk, disability, disease activity, and higher quality of life (QoL) [135]. Healthier diet scores also associated with better mental, physical, and total QoL, improved cognitive function, lower depression, anxiety, and pain scores, and fewer cognition, vision, and bowel symptoms [136138].

Numerous studies have identified foods and food groups that worsen various clinical outcomes of MS. Higher consumption of beef, butter, pastries, and sweets associated with increased risk [130,131], whereas not consuming meat correlated with reduced disability progression in pwMS [139]. Diets enriched in added sugars and processed foods corresponded with higher burden of metabolic risk factors including abdominal obesity and reduced circulating high-density lipoprotein concentrations among pwMS [140]. Case-control studies corroborated that pro-inflammatory and low antioxidant-containing diets increased MS risk [129,141144].

4.3 Dietary patterns that improve MS

Various dietary patterns have been tested in preliminary trials including the low-fat, Mediterranean, Paleolithic, ketogenic, caloric restriction, fast-mimicking, and time-restricted feeding diets. In the 1950s, Dr. Roy Swank reported an association between saturated fat intake and risk of MS [145]. He recommended a low-saturated fat diet to his patients [146], followed them for up to 50 years [147], and observed that patients who consumed the least amount of saturated fats were less likely to have disease exacerbations, more likely to retain ambulation, and had reduced risk of mortality [148,149]. Further support for reducing dietary saturated fat comes from a multi-center prospective cohort study in pediatric individuals with MS. This study reported that each 10% increase in energy intake from fat, particularly saturated fat, increased the risk of relapse and each additional one cup equivalent of vegetables decreased relapse risk [150]. Low-fat diets have also been reported to reduce fatigue and improve QoL [151153]. Curiously, a randomized controlled trial found that a plant-based, low-fat diet did not affect magnetic resonance imaging (MRI) outcomes, relapses, or expanded disability status scale (EDSS) scores over 12 months; however, the control group included an exercise intervention, which confounds this finding [153].

Numerous MS studies have reported positive impacts of the Mediterranean diet and the Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND). These dietary patterns and other diets with high intake of fruits and vegetables associated with reduced risk of MS [130,132,142].The Mediterranean diet reduced fatigue and improved QoL in several trials [154159], and in a 6-month randomized controlled trial, reduced EDSS progression compared with controls among women with MS [155]. Adherence to the MIND diet associated with thalamic volume in a cross-sectional study among 180 individuals with early MS [133]. Corroborating data from a 5-year longitudinal case-control study among 175 MS cases and 42 controls showed that the AHA Healthy Heart Score diet component inversely associated with 5-year T2 lesion volume accrual [160].

Multiple dietary patterns with features of ancestral eating patterns have gained attention for pwMS. A multimodal intervention that included a Paleolithic diet led to improved fatigue, QoL, gait, mood, and cholesterol profiles in a single-arm trial [161165]. Additionally, two randomized controlled trials and one parallel-arm trial confirmed that a modified Paleolithic diet reduces fatigue and improves QoL [151,166,167]. Ketogenic diets yielded similar improvements [96,167169]. This form of carbohydrate restriction normalizes the colonic microbiome, reduces oxidative stress, inflammation, and serum neurofilament light chain (sNFL) [170172]. In addition, several calorie-restricted and fasting strategies gave mixed results in preliminary trials with adherence being a primary barrier to conclusive outcomes [173175].

4.4 Conflicting findings of dietary clinical trials

Not all trials and prospective studies have yielded clear findings to inform dietary guidelines. Most notably, two prospective cohort studies among 56,867 Danish adults and 185,000 US women found no association between diet quality and MS risk [176,177]. The infrequent incidence of MS incidence in both studies (0.2%) may underlie these null findings. In addition, cross-sectional associations of diet quality can be confounded by disability burdens that preclude acquiring and preparing healthy foods [178]. In fact, recent meta-analysis and network meta-analysis of randomized dietary intervention trials found consistent positive effects of several dietary interventions (eg Paleolithic and Mediterranean) on fatigue and QoL; however, due to the small sample size and methodological issues among the preliminary trials, the evidence had limited reliability [179,180].

5. Potential intersections of dietary interventions and MS-ADON

In the absence of clinically-validated nutritional recommendations to advance specific diets that are effective and safe for mitigating MS-ADON, the following section provides dietary guidelines that we posit may prove efficacious based on the multiple mechanisms putatively involved with this ocular sequel: (1) Diets stressing whole foods while reducing or eliminating ultra-processed foods laden with added sugars, additives, hydrogenated fats, and sodium. This recommendation stems from the findings that ultra-processed food consumption associates with increased likelihood of an FDE [124,181] and with increased MS severity [125]. Furthermore, an abundance of dietary glucose promotes TH17 cell differentiation [182] and auto-reactive TH17 cells are implicated as primary drivers of ADON (eg [44,46]). (2) Diets enriched in ω3 anti-inflammatory FAs and limited in ω6 pro-inflammatory FAs. In alignment with the foundational role of inflammation in MS, the types of dietary fats consumed can either positively or negatively impact systemic states of inflammation and gut integrity [99]. For example, ω3 anti-inflammatory FAs, which are enriched in foods like mackerel, salmon, tuna, walnuts, and flaxseed oil, or obtained through supplements, provide precursors for increasing the synthesis of specialized pro-resolving lipid mediators including the resolvins and other endogenous factors that mitigate inflammation. Notably, a meta-analysis of observational studies found that fish consumption is associated with a 23% decreased risk of MS [183]. In contrast, arachidonic acid, select other ω6 FAs, and saturated fats (eg [146,147,184]) should be limited as these lipids fuel the synthesis of pro-inflammatory leukotrienes and prostaglandins [185,186]. Additionally, dietary short-chain FAs, such as propionic acid, have shown benefit in reducing EAE disease severity, whereas dietary long-chain FAs such as lauric acid and palmitic acid (present in coconut oil, palm oil, and soybean oil) exacerbate severity [99]. (3) Diets containing sufficient fermentable and non-fermentable fiber to support a healthy gut microbiome. The commensal microbiota and their collective metabolites are primary regulators of immune cell function, health, and autoimmunity, particularly in the context of T cell-driven autoimmunity in tissues distant from the gut, including the eye [187,188]. Dietary fiber is essential for maintaining a healthy commensal microbiota [189191] and low consumption of sufficient fiber has been associated with increased FDE incidence [127], whereas higher dietary fiber correlates with reduced motor deficits, optic neuritis, and visual acuity loss in EAE mice [117,192,193] and overall better health outcomes [128]. (4) Dietary patterns that reduce biomarkers of MS disease severity. A recent study [171] showed that 6 months of an adapted ketogenic diet (ie, average daily intake of >160 g fat [ω6 vs ω3 ratio 2:1], <50 g carbohydrates, and average protein intake ≤100 g/day) reduced sNFL, a recognized biomarker of MS severity [194197]. Additional clinical and animal evidence support the efficacy of ketogenic diets and other approaches (eg, fast-mimicking diets and intermittent fasting) that induce starvation responses as a common mechanism to ameliorate ADON (eg, [117] and other diseases [reviewed in [198]]). Recent work has shown that a ketogenic diet can decrease pro-inflammatory TH17 cells in the lamina propria, a mucosal layer of the small intestine, and modulate the gut microbiome [199]. However, these strategies may need to be used intermittently (eg, in response to ADON flare-ups) considering long-term compliance is challenging [200,201]. Conflicting results regarding the cardiometabolic impacts of chronic nutritional ketosis also need to be resolved for this approach to gain wider acceptance [202,203]. (5) Identification and elimination of potential autoimmune exacerbating dietary antigens. Sensitivity to dietary antigens is estimated to affect 15% to 20% of the population [204]. Among people with sensitivities, dietary antigens can drive inflammation of the intestinal mucosal layer. However, the link between dietary antigens and MS remains elusive. The modified Paleolithic elimination diet has shown efficacy in clinical trials (eg, [151,161,162,165,166,205,206]) and is based on consuming whole foods while limiting simple sugars, pro-inflammatory fats, as well as gluten, lactose, legumes, nightshades, soy, and whole eggs. pwMS can further optimize this approach by re-introducing select components to identify specific dietary triggers of symptom flare-ups. Additional dietary approaches eliminating antigen-rich foods that presumably drive inflammatory responses have also elicited favorable impacts on MS symptoms (eg, [157,207,208]).

6. Conclusions

Disease-specific dietary recommendations are highly desired by pwMS [209212]. In addition to identifying efficacious dietary strategies, it will be important to develop, in parallel, mechanisms to facilitate adherence. Fortunately, because numerous diets and nutritional approaches have shown benefit in preliminary studies, therapeutic diets may be customizable for each pwMS by taking into account personal and familial dietary preferences, culture, costs, health and metabolic status, and logistical factors such as the ability to acquire and prepare food. The availability of medical foods combined with delivery services will further facilitate access to customized diets, and continuous remote care involving health coaching and wearables (eg continuous glucose monitors) are anticipated to promote adherence.

Word limitations precluded discussing in appropriate detail many diet-related topics relevant to autoimmune demyelination. We refer interested readers to publications covering the central contributions to autoimmunity of FAs [14,213], gut dysbiosis, and microbiome (eg, [214220], and micronutrients [11,206,221225]).

Conflicts of interest

The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: Dr Terry Wahls has equity interest in the following companies: Terry Wahls LLC; TZ Press LLC; The Wahls Institute, PLC; FBB Biomed Inc; and the website http://www.terry-wahls.com. She also owns the copyright to the books Minding My Mitochondria (2nd Edition) and The Wahls Protocol, The Wahls Protocol Cooking for Life, and the trademarks The Wahls Protocol and Wahls diet, Wahls Paleo diet, and Wahls Paleo Plus diets. She has completed grant funding from the National Multiple Sclerosis Society for the Dietary Approaches to Treating Multiple Sclerosis Related Fatigue Study. She has financial relationships with BioCeuticals Ltd., MCG Health LLC, Vibrant America LLC, Standard Process Inc., MasterHealth Technologies Inc., Foogal Inc., Genova Diagnostics Inc., and the Institute for Functional Medicine. She receives royalty payments from Penguin Random House. Dr Wahls has conflict of interest management plans in place with the University of Iowa and the Iowa City Veteran’s Affairs Medical Center. All other authors report no personal or financial conflicts of interest in this work.

Funding

The authors additionally acknowledge the financial support from the Presbyterian Health Foundation (to S.M.P.). T.J.T. is supported by the Carter Chapman Shreve Family Foundation and the Carter Chapman Shreve Fellowship Fund for diet and lifestyle research conducted by the Wahls Research team at the University of Iowa.

Acknowledgments

The authors acknowledge the support from the National Eye Institute of the National Institutes of Health under Award Number R01EY033782 (to S.M.P.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. In-kind support was provided by the University of Iowa College of Public Health Preventive Intervention Center. Special thanks to KS Plafker for assistance with literature searches and editing

Footnotes

These authors contributed equally to this work.

Data sharing not applicable to this article as no datasets were generated or analyzed during the current study.

How to cite this article: Plafker SM, Titcomb T, Zyla-Jackson K, et al. Overview of diet and autoimmune demyelinating optic neuritis: a narrative review. Immunometabolism. 2023;5(2):e00022. doi: 10.1097/IN9.0000000000000022.

References

  • [1].Bach JF. The hygiene hypothesis in autoimmunity: the role of pathogens and commensals. Nat Rev Immunol. 2018;18(2):105–20. [DOI] [PubMed] [Google Scholar]
  • [2].Koch-Henriksen N, Sorensen PS. The changing demographic pattern of multiple sclerosis epidemiology. Lancet Neurol. 2010;9(5):520–32. [DOI] [PubMed] [Google Scholar]
  • [3].Dinse GE, Parks CG, Weinberg CR, et al. Increasing prevalence of antinuclear antibodies in the United States. Arthritis Rheumatol. 2020;72(6):1026–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Louzada ML, Martins AP, Canella DS, et al. Impact of ultra-processed foods on micronutrient content in the Brazilian diet. Rev Saude Publica. 2015;49:45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Martini D, Godos J, Bonaccio M, et al. Ultra-processed foods and nutritional dietary profile: a meta-analysis of Nationally representative samples. Nutrients. 2021;13(10):33903390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Bolte LA, Vich Vila A, Imhann F, et al. Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut. 2021;70(7):1287–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Shi Z. Gut microbiota: an important link between western diet and chronic diseases. Nutrients. 2019;11(10):22872287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Zinocker MK, Lindseth IA. The Western diet-microbiome-host interaction and its role in metabolic disease. Nutrients. 2018;10(3):365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Martel J, Chang SH, Ko YF, et al. Gut barrier disruption and chronic disease. Trends Endocrinol Metab. 2022;33(4):247–65. [DOI] [PubMed] [Google Scholar]
  • [10].Martinelli V, Albanese M, Altieri M, et al. Gut-oriented interventions in patients with multiple sclerosis: fact or fiction? Eur Rev Med Pharmacol Sci. 2022;26(3):935–46. [DOI] [PubMed] [Google Scholar]
  • [11].Titcomb TJ, Bisht B, Moore DD, et al. Eating pattern and nutritional risks among people with multiple sclerosis following a modified paleolithic diet. Nutrients. 2020;12(6):1844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Wahls TL, Chenard CA, Snetselaar LG. Review of two popular eating plans within the multiple sclerosis community: low saturated fat and modified paleolithic. Nutrients. 2019;11(2):352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Winiarska-Mieczan A, Tomaszewska E, Donaldson J, et al. The role of nutritional factors in the modulation of the composition of the gut microbiota in people with autoimmune diabetes. Nutrients. 2022;14(12):24982498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Yu H, Bai S, Hao Y, et al. Fatty acids role in multiple sclerosis as “metabokines”. J Neuroinflammation. 2022;19(1):157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nat Rev Immunol. 2014;14(10):667–85. [DOI] [PubMed] [Google Scholar]
  • [16].Balcer LJ. Clinical practice. Optic neuritis. N Engl J Med. 2006;354(12):1273–80. [DOI] [PubMed] [Google Scholar]
  • [17].Jarius S, Paul F, Weinshenker BG, et al. Neuromyelitis optica. Nat Rev Dis Primers. 2020;6(1):85. [DOI] [PubMed] [Google Scholar]
  • [18].Reindl M, Rostasy K. MOG antibody-associated diseases. Neurol Neuroimmunol Neuroinflammation. 2015;2(1):e60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Reindl M, Waters P. Myelin oligodendrocyte glycoprotein antibodies in neurological disease. Nat Rev Neurol. 2019;15(2):89–102. [DOI] [PubMed] [Google Scholar]
  • [20].Sechi E, Cacciaguerra L, Chen JJ, et al. Myelin Oligodendrocyte Glycoprotein Antibody-Associated Disease (MOGAD): a review of clinical and mri features, diagnosis, and management. Front Neurol. 2022;13:885218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Kale N. Optic neuritis as an early sign of multiple sclerosis. Eye Brain. 2016;8:195–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Salter AR, Tyry T, Vollmer T, et al. “Seeing” in NARCOMS: a look at vision-related quality of life in the NARCOMS registry. Mult Scler. 2013;19(7):953–60. [DOI] [PubMed] [Google Scholar]
  • [23].Sorensen TL, Frederiksen JL, Bronnum-Hansen H, et al. Optic neuritis as onset manifestation of multiple sclerosis: a nationwide, long-term survey. Neurology. 1999;53(3):473–8. [DOI] [PubMed] [Google Scholar]
  • [24].Jarius S, Ruprecht K, Kleiter I, et al. ; in cooperation with the Neuromyelitis Optica Study Group (NEMOS). MOG-IgG in NMO and related disorders: a multicenter study of 50 patients. Part 1: Frequency, syndrome specificity, influence of disease activity, long-term course, association with AQP4-IgG, and origin. J Neuroinflammation. 2016;13(1):279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Jarius S, Ruprecht K, Wildemann B, et al. Contrasting disease patterns in seropositive and seronegative neuromyelitis optica: a multicentre study of 175 patients. J Neuroinflammation. 2012;9:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Messias K, Marques VD, Messias A. Myelin oligodendrocyte glycoprotein antibody-associated optic neuritis: an update. Arq Bras Oftalmol. 2023;86(1):83–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Wilhelm H, Schabet M. The diagnosis and treatment of optic neuritis. Dtsch Arztebl Int. 2015;112(37):616–25; quiz 626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Bennett JL, Costello F, Chen JJ, et al. Optic neuritis and autoimmune optic neuropathies: advances in diagnosis and treatment. Lancet Neurol. 2023;22(1):89–100. [DOI] [PubMed] [Google Scholar]
  • [29].Horton L, Bennett JL. Acute management of optic neuritis: an evolving paradigm. J Neuroophthalmol. 2018;38(3):358–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Touma L, Muccilli A. Diagnosis and management of central nervous system demyelinating disorders. Neurol Clin. 2022;40(1):113–31. [DOI] [PubMed] [Google Scholar]
  • [31].Hacohen Y, Wong YY, Lechner C, et al. Disease course and treatment responses in children with relapsing myelin oligodendrocyte glycoprotein antibody-associated disease. JAMA Neurol. 2018;75(4):478–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Wildemann B, Jarius S, Schwarz A, et al. Failure of alemtuzumab therapy to control MOG encephalomyelitis. Neurology. 2017;89(2):207–9. [DOI] [PubMed] [Google Scholar]
  • [33].Glatigny S, Bettelli E. Experimental autoimmune encephalomyelitis (EAE) as animal models of multiple sclerosis (MS). Cold Spring Harb Perspect Med. 2018;8(11):a028977a028977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Bettelli E, Pagany M, Weiner HL, et al. Myelin oligodendrocyte glycoprotein-specific T cell receptor transgenic mice develop spontaneous autoimmune optic neuritis. J Exp Med. 2003;197(9):1073–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Krishnamoorthy G, Lassmann H, Wekerle H, et al. Spontaneous opticospinal encephalomyelitis in a double-transgenic mouse model of autoimmune T cell/B cell cooperation. J Clin Invest. 2006;116(9):2385–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Regen T, Isaac S, Amorim A, et al. IL-17 controls central nervous system autoimmunity through the intestinal microbiome. Sci Immunol. 2021;6(56):eaaz6563. [DOI] [PubMed] [Google Scholar]
  • [37].Bellone M, Brevi A, Huber S. Microbiota-propelled T helper 17 cells in inflammatory diseases and cancer. Microbiol Mol Biol Rev. 2020;84(2):e00064–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Omenetti S, Bussi C, Metidji A, et al. The intestine harbors functionally distinct homeostatic tissue-resident and inflammatory Th17 cells. Immunity. 2019;51(1):77–89.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Matusevicius D, Kivisakk P, He B, et al. Interleukin-17 mRNA expression in blood and CSF mononuclear cells is augmented in multiple sclerosis. Mult Scler. 1999;5(2):101–4. [DOI] [PubMed] [Google Scholar]
  • [40].Cua DJ, Tato CM. Innate IL-17-producing cells: the sentinels of the immune system. Nat Rev Immunol. 2010;10(7):479–89. [DOI] [PubMed] [Google Scholar]
  • [41].Hirota K, Duarte JH, Veldhoen M, et al. Fate mapping of IL-17-producing T cells in inflammatory responses. Nat Immunol. 2011;12(3):255–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Komiyama Y, Nakae S, Matsuki T, et al. IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis. J Immunol. 2006;177(1):566–73. [DOI] [PubMed] [Google Scholar]
  • [43].Sutton CE, Lalor SJ, Sweeney CM, et al. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity. 2009;31(2):331–41. [DOI] [PubMed] [Google Scholar]
  • [44].Larabee CM, Hu Y, Desai S, et al. Myelin-specific Th17 cells induce severe relapsing optic neuritis with irreversible loss of retinal ganglion cells in C57BL/6 mice. Mol Vis. 2016;22:332–41. [PMC free article] [PubMed] [Google Scholar]
  • [45].Kaskow BJ, Baecher-Allan C. Effector T cells in multiple sclerosis. Cold Spring Harb Perspect Med. 2018;8(4):a029025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Knier B, Rothhammer V, Heink S, et al. Neutralizing IL-17 protects the optic nerve from autoimmune pathology and prevents retinal nerve fiber layer atrophy during experimental autoimmune encephalomyelitis. J Autoimmun. 2015;56:34–44. [DOI] [PubMed] [Google Scholar]
  • [47].Kang Z, Wang C, Zepp J, et al. Act1 mediates IL-17-induced EAE pathogenesis selectively in NG2+ glial cells. Nat Neurosci. 2013;16(10):1401–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Denic A, Wootla B, Rodriguez M. CD8(+) T cells in multiple sclerosis. Expert Opin Ther Targets. 2013;17(9):1053–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Friese MA, Fugger L. Pathogenic CD8(+) T cells in multiple sclerosis. Ann Neurol. 2009;66(2):132–41. [DOI] [PubMed] [Google Scholar]
  • [50].Brynedal B, Duvefelt K, Jonasdottir G, et al. HLA-A confers an HLA-DRB1 independent influence on the risk of multiple sclerosis. PLoS One. 2007;2(7):e664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Fogdell-Hahn A, Ligers A, Gronning M, et al. Multiple sclerosis: a modifying influence of HLA class I genes in an HLA class II associated autoimmune disease. Tissue Antigens. 2000;55(2):140–8. [DOI] [PubMed] [Google Scholar]
  • [52].Harbo HF, Lie BA, Sawcer S, et al. Genes in the HLA class I region may contribute to the HLA class II-associated genetic susceptibility to multiple sclerosis. Tissue Antigens. 2004;63(3):237–47. [DOI] [PubMed] [Google Scholar]
  • [53].Sawcer S, Hellenthal G, Pirinen M, et al. ; International Multiple Sclerosis Genetics Consortium. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011;476(7359):214–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Jilek S, Schluep M, Rossetti AO, et al. CSF enrichment of highly differentiated CD8+ T cells in early multiple sclerosis. Clin Immunol. 2007;123(1):105–13. [DOI] [PubMed] [Google Scholar]
  • [55].Hauser SL, Bhan AK, Gilles F, et al. Immunohistochemical analysis of the cellular infiltrate in multiple sclerosis lesions. Ann Neurol. 1986;19(6):578–87. [DOI] [PubMed] [Google Scholar]
  • [56].Traugott U, Reinherz EL, Raine CS. Multiple sclerosis. Distribution of T cells, T cell subsets and Ia-positive macrophages in lesions of different ages. J Neuroimmunol. 1983;4(3):201–21. [DOI] [PubMed] [Google Scholar]
  • [57].Babbe H, Roers A, Waisman A, et al. Clonal expansions of CD8(+) T cells dominate the T cell infiltrate in active multiple sclerosis lesions as shown by micromanipulation and single cell polymerase chain reaction. J Exp Med. 2000;192(3):393–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Tsakiri A, Kjaersgaard E, Grigoriadis N, et al. Effector and regulatory T cells in patients with acute optic neuritis. Neuroimmunomodulation. 2012;19(2):111–20. [DOI] [PubMed] [Google Scholar]
  • [59].Wang HH, Dai YQ, Qiu W, et al. Interleukin-17-secreting T cells in neuromyelitis optica and multiple sclerosis during relapse. J Clin Neurosci. 2011;18(10):1313–7. [DOI] [PubMed] [Google Scholar]
  • [60].Shimizu Y, Ota K, Kubo S, et al. Association of Th1/Th2-related chemokine receptors in peripheral T cells with disease activity in patients with multiple sclerosis and neuromyelitis optica. Eur Neurol. 2011;66(2):91–7. [DOI] [PubMed] [Google Scholar]
  • [61].Shi Z, Qiu Y, Zhao Z, et al. CD8(+) T cell subpopulations and pro-inflammatory cytokines in neuromyelitis optica spectrum disorder. Ann Clin Transl Neurol. 2021;8(1):43–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Brambilla R, Dvoriantchikova G, Barakat D, et al. Transgenic inhibition of astroglial NF-kappaB protects from optic nerve damage and retinal ganglion cell loss in experimental optic neuritis. J Neuroinflammation. 2012;9:213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Chun BY, Kim JH, Nam Y, et al. Pathological Involvement of Astrocyte-Derived Lipocalin-2 in the Demyelinating Optic Neuritis. Invest Ophthalmol Vis Sci. 2015;56(6):3691–8. [DOI] [PubMed] [Google Scholar]
  • [64].Tassoni A, Farkhondeh V, Itoh Y, et al. The astrocyte transcriptome in EAE optic neuritis shows complement activation and reveals a sex difference in astrocytic C3 expression. Sci Rep. 2019;9(1):10010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Nataf S, Carroll SL, Wetsel RA, et al. Attenuation of experimental autoimmune demyelination in complement-deficient mice. J Immunol. 2000;165(10):5867–73. [DOI] [PubMed] [Google Scholar]
  • [66].Szalai AJ, Hu X, Adams JE, et al. Complement in experimental autoimmune encephalomyelitis revisited: C3 is required for development of maximal disease. Mol Immunol. 2007;44(12):3132–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Ingram G, Loveless S, Howell OW, et al. Complement activation in multiple sclerosis plaques: an immunohistochemical analysis. Acta Neuropathol Commun. 2014;2:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Godwin CR, Anders JJ, Cheng L, et al. Targeting cholesterol homeostasis improves recovery in experimental optic neuritis. Biomolecules. 2022;12(10):14371437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Achiron A, Hecht I, Abayev L, et al. B-cell related biomarkers associated with severity of the first demyelinating event of acute optic neuritis. Eye (Lond). 2020;34(5):954–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Feldman A, Gurevich M, Huna-Baron R, et al. The role of B cells in the early onset of the first demyelinating event of acute optic neuritis. Invest Ophthalmol Vis Sci. 2015;56(2):1349–56. [DOI] [PubMed] [Google Scholar]
  • [71].Lundqvist S, Modvig S, Fischer EA, et al. Frequency and immunophenotype of IL10-producing regulatory B cells in optic neuritis. Immunology. 2019;156(3):259–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Fernandez Blanco L, Marzin M, Leistra A, et al. Immunopathology of the optic nerve in multiple sclerosis. Clin Exp Immunol. 2022;209(2):236–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Trebst C, Sorensen TL, Kivisakk P, et al. CCR1+/CCR5+ mononuclear phagocytes accumulate in the central nervous system of patients with multiple sclerosis. Am J Pathol. 2001;159(5):1701–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Vogel DY, Vereyken EJ, Glim JE, et al. Macrophages in inflammatory multiple sclerosis lesions have an intermediate activation status. J Neuroinflammation. 2013;10:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Zhang Z, Zhang ZY, Schittenhelm J, et al. Parenchymal accumulation of CD163+ macrophages/microglia in multiple sclerosis brains. J Neuroimmunol. 2011;237(1-2):73–9. [DOI] [PubMed] [Google Scholar]
  • [76].Kouwenhoven M, Teleshova N, Ozenci V, et al. Monocytes in multiple sclerosis: phenotype and cytokine profile. J Neuroimmunol. 2001;112(1-2):197–205. [DOI] [PubMed] [Google Scholar]
  • [77].Gramlich OW, Joachim SC, Gottschling PF, et al. Ophthalmopathology in rats with MBP-induced experimental autoimmune encephalomyelitis. Graefes Arch Clin Exp Ophthalmol. 2011;249(7):1009–20. [DOI] [PubMed] [Google Scholar]
  • [78].Heppner FL, Greter M, Marino D, et al. Experimental autoimmune encephalomyelitis repressed by microglial paralysis. Nat Med. 2005;11(2):146–52. [DOI] [PubMed] [Google Scholar]
  • [79].Manogaran P, Samardzija M, Schad AN, et al. Retinal pathology in experimental optic neuritis is characterized by retrograde degeneration and gliosis. Acta Neuropathol Commun. 2019;7(1):116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Ponomarev ED, Shriver LP, Maresz K, et al. Microglial cell activation and proliferation precedes the onset of CNS autoimmunity. J Neurosci Res. 2005;81(3):374–89. [DOI] [PubMed] [Google Scholar]
  • [81].Yamasaki R, Lu H, Butovsky O, et al. Differential roles of microglia and monocytes in the inflamed central nervous system. J Exp Med. 2014;211(8):1533–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Horstmann L, Kuehn S, Pedreiturria X, et al. Microglia response in retina and optic nerve in chronic experimental autoimmune encephalomyelitis. J Neuroimmunol. 2016;298:32–41. [DOI] [PubMed] [Google Scholar]
  • [83].Horstmann L, Schmid H, Heinen AP, et al. Inflammatory demyelination induces glia alterations and ganglion cell loss in the retina of an experimental autoimmune encephalomyelitis model. J Neuroinflammation. 2013;10:120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Savarin C, Hinton DR, Valentin-Torres A, et al. Astrocyte response to IFN-gamma limits IL-6-mediated microglia activation and progressive autoimmune encephalomyelitis. J Neuroinflammation. 2015;12:79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Tran EH, Hoekstra K, van Rooijen N, et al. Immune invasion of the central nervous system parenchyma and experimental allergic encephalomyelitis, but not leukocyte extravasation from blood, are prevented in macrophage-depleted mice. J Immunol. 1998;161(7):3767–75. [PubMed] [Google Scholar]
  • [86].Fan X, Zhang H, Cheng Y, et al. Double roles of macrophages in human neuroimmune diseases and their animal models. Mediators Inflamm. 2016;2016:8489251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Liu F, Alfarhan M, Baker L, et al. Treatment with MDL 72527 ameliorated clinical symptoms, retinal ganglion cell loss, optic nerve inflammation, and improved visual acuity in an experimental model of multiple sclerosis. Cells. 2022;11(24):41004100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Candadai AA, Liu F, Fouda AY, et al. Deletion of arginase 2 attenuates neuroinflammation in an experimental model of optic neuritis. PLoS One. 2021;16(3):e0247901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Guo J, Li B, Wang J, et al. Protective effect and mechanism of nicotinamide adenine dinucleotide against optic neuritis in mice with experimental autoimmune encephalomyelitis. Int Immunopharmacol. 2021;98:107846. [DOI] [PubMed] [Google Scholar]
  • [90].Liu C, Li Y, Yu J, et al. Targeting the shift from M1 to M2 macrophages in experimental autoimmune encephalomyelitis mice treated with fasudil. PLoS One. 2013;8(2):e54841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Porcheray F, Viaud S, Rimaniol AC, et al. Macrophage activation switching: an asset for the resolution of inflammation. Clin Exp Immunol. 2005;142(3):481–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Liu Y, Given KS, Owens GP, et al. Distinct patterns of glia repair and remyelination in antibody-mediated demyelination models of multiple sclerosis and neuromyelitis optica. Glia. 2018;66(12):2575–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Kigerl KA, Gensel JC, Ankeny DP, et al. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci. 2009;29(43):13435–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Olesen MN, Soelberg K, Debrabant B, et al. Cerebrospinal fluid biomarkers for predicting development of multiple sclerosis in acute optic neuritis: a population-based prospective cohort study. J Neuroinflammation. 2019;16(1):59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Bai M, Wang Y, Han R, et al. Intermittent caloric restriction with a modified fasting-mimicking diet ameliorates autoimmunity and promotes recovery in a mouse model of multiple sclerosis. J Nutr Biochem. 2021;87:108493. [DOI] [PubMed] [Google Scholar]
  • [96].Choi IY, Piccio L, Childress P, et al. A Diet Mimicking Fasting Promotes Regeneration and Reduces Autoimmunity and Multiple Sclerosis Symptoms. Cell Rep. 2016;15(10):2136–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Cignarella F, Cantoni C, Ghezzi L, et al. Intermittent Fasting Confers Protection in CNS Autoimmunity by Altering the Gut Microbiota. Cell Metab. 2018;27(6):1222–1235.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Dworkin RH, Bates D, Millar JH, et al. Linoleic acid and multiple sclerosis: a reanalysis of three double-blind trials. Neurology. 1984;34(11):1441–5. [DOI] [PubMed] [Google Scholar]
  • [99].Haghikia A, Jorg S, Duscha A, et al. Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine. Immunity. 2015;43(4):817–29. [DOI] [PubMed] [Google Scholar]
  • [100].Harbige LS, Layward L, Morris-Downes MM, et al. 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. 2000;122(3):445–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Harbige LS, Yeatman N, Amor S, et al. Prevention of experimental autoimmune encephalomyelitis in Lewis rats by a novel fungal source of gamma-linolenic acid. Br J Nutr. 1995;74(5):701–15. [DOI] [PubMed] [Google Scholar]
  • [102].Kafami L, Raza M, Razavi A, et al. Intermittent feeding attenuates clinical course of experimental autoimmune encephalomyelitis in C57BL/6 mice. Avicenna J Med Biotechnol. 2010;2(1):47–52. [PMC free article] [PubMed] [Google Scholar]
  • [103].Mancera P, Wappenhans B, Cordobilla B, et al. Natural docosahexaenoic acid in the triglyceride form attenuates in vitro microglial activation and ameliorates autoimmune encephalomyelitis in mice. Nutrients. 2017;9(7):681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Meade CJ, Mertin J, Sheena J, et al. Reduction by linoleic acid of the severity of experimental allergic encephalomyelitis in the guinea pig. J Neurol Sci. 1978;35(2-3):291–308. [DOI] [PubMed] [Google Scholar]
  • [105].Na SY, Janakiraman M, Leliavski A, et al. High-salt diet suppresses autoimmune demyelination by regulating the blood-brain barrier permeability. Proc Natl Acad Sci U S A. 2021;118(12):e2025944118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Piccio L, Stark JL, Cross AH. Chronic calorie restriction attenuates experimental autoimmune encephalomyelitis. J Leukoc Biol. 2008;84(4):940–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Razeghi Jahromi S, Ghaemi A, Alizadeh A, et al. Effects of intermittent fasting on experimental autoimune encephalomyelitis in C57BL/6 mice. Iran J Allergy Asthma Immunol. 2016;15(3):212–9. [PubMed] [Google Scholar]
  • [108].Unoda K, Doi Y, Nakajima H, et al. Eicosapentaenoic acid (EPA) induces peroxisome proliferator-activated receptors and ameliorates experimental autoimmune encephalomyelitis. J Neuroimmunol. 2013;256(1-2):7–12. [DOI] [PubMed] [Google Scholar]
  • [109].Valburg C, Sonti A, Stern JN, et al. Dietary factors in experimental autoimmune encephalomyelitis and multiple sclerosis: a comprehensive review. Mult Scler. 2021;27(4):494–502. [DOI] [PubMed] [Google Scholar]
  • [110].Wilck N, Matus MG, Kearney SM, et al. Salt-responsive gut commensal modulates TH17 axis and disease. Nature. 2017;551(7682):585–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Dal Monte M, Cammalleri M, Locri F, et al. Fatty acids dietary supplements exert anti-inflammatory action and limit ganglion cell degeneration in the retina of the eae mouse model of multiple sclerosis. Nutrients. 2018;10(3):325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Locri F, Cammalleri M, Pini A, et al. Further evidence on efficacy of diet supplementation with fatty acids in ocular pathologies: insights from the EAE model of optic neuritis. Nutrients. 2018;10(10):14471447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Fleck AK, Hucke S, Teipel F, et al. Dietary conjugated linoleic acid links reduced intestinal inflammation to amelioration of CNS autoimmunity. Brain. 2021;144(4):1152–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Liu Q, Li H, Yang J, et al. Valproic acid attenuates inflammation of optic nerve and apoptosis of retinal ganglion cells in a rat model of optic neuritis. Biomed Pharmacother. 2017;96:1363–70. [DOI] [PubMed] [Google Scholar]
  • [115].Chaudhary P, Marracci G, Yu X, et al. Lipoic acid decreases inflammation and confers neuroprotection in experimental autoimmune optic neuritis. J Neuroimmunol. 2011;233(1-2):90–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Dietrich M, Helling N, Hilla A, et al. Early alpha-lipoic acid therapy protects from degeneration of the inner retinal layers and vision loss in an experimental autoimmune encephalomyelitis-optic neuritis model. J Neuroinflammation. 2018;15(1):71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Zyla-Jackson WD, Plafker KS, Kovats S, et al. Dietary protection against the visual and motor deficits induced by experimental autoimmune encephalomyelitis. Front Neurol. 2023;14:1113954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Black LJ, Bowe GS, Pereira G, et al. ; Ausimmune Investigator Group. Higher non-processed red meat consumption is associated with a reduced risk of central nervous system demyelination. Front Neurol. 2019;10:125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Black LJ, Zhao Y, Peng YC, et al. ; Ausimmune Investigator Group. Higher fish consumption and lower risk of central nervous system demyelination. Eur J Clin Nutr. 2020;74(5):818–24. [DOI] [PubMed] [Google Scholar]
  • [120].Dieu DYR, Dunlop E, Daly A, et al. Total dairy consumption is not associated with likelihood of a first clinical diagnosis of central nervous system demyelination. Front Neurol. 2022;13:888559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Black LJ, Baker K, Ponsonby AL, et al. ; Ausimmune Investigator Group. A higher mediterranean diet score, including unprocessed red meat, is associated with reduced risk of central nervous system demyelination in a case-control study of australian adults. J Nutr. 2019;149(8):1385–92. [DOI] [PubMed] [Google Scholar]
  • [122].Black LJ, Rowley C, Sherriff J, et al. A healthy dietary pattern associates with a lower risk of a first clinical diagnosis of central nervous system demyelination. Mult Scler. 2019;25(11):1514–25. [DOI] [PubMed] [Google Scholar]
  • [123].Shivappa N, Steck SE, Hurley TG, et al. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Mannino A, Lithander FE, Dunlop E, et al. ; Ausimmune Investigator Group. A proinflammatory diet is associated with an increased likelihood of first clinical diagnosis of central nervous system demyelination in women. Mult Scler Relat Disord. 2022;57:103428. [DOI] [PubMed] [Google Scholar]
  • [125].Guglielmetti M, Grosso G, Ferraris C, et al. Ultra-processed foods consumption is associated with multiple sclerosis severity. Front Neurol. 2023;14:1086720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Hajianfar H, Mirmossayeb O, Mollaghasemi N, et al. Association between dietary inflammatory index and risk of demyelinating autoimmune diseases. Int J Vitam Nutr Res. 2022. doi: 10.1024/0300-9831/a000754. [DOI] [PubMed] [Google Scholar]
  • [127].Cavalla P, Golzio P, Maietta D, et al. Dietary habits, nutritional status and risk of a first demyelinating event: an incident case-control study in a southern European cohort. Neurol Sci. 2022;43(7):4373–80. [DOI] [PubMed] [Google Scholar]
  • [128].Marck CH, Probst Y, Chen J, et al. Dietary patterns and associations with health outcomes in Australian people with multiple sclerosis. Eur J Clin Nutr. 2021;75(10):1506–14. [DOI] [PubMed] [Google Scholar]
  • [129].Abdollahpour I, Nedjat S, Salimi Y, et al. The role of dietary antioxidant index and index of nutritional quality in MS onset: finding from an Iranian population-based incident case-control study. Nutr Neurosci. 2022;25(2):379–86. [DOI] [PubMed] [Google Scholar]
  • [130].Noormohammadi M, Ghorbani Z, Naser Moghadasi A, et al. MIND diet adherence might be associated with a reduced odds of multiple sclerosis: results from a case-control study. Neurol Ther. 2022;11(1):397–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Pekmezovic TD, Tepavcevic DBK, Mesaros ST, et al. Food and dietary patterns and multiple sclerosis: a case-control study in Belgrade (Serbia). Ital J Public Health. 2012;6:81–7. [Google Scholar]
  • [132].Sedaghat F, Jessri M, Behrooz M, et al. Mediterranean diet adherence and risk of multiple sclerosis: a case-control study. Asia Pac J Clin Nutr. 2016;25(2):377–84. [DOI] [PubMed] [Google Scholar]
  • [133].Katz Sand IB, Fitzgerald KC, Gu Y, et al. Dietary factors and MRI metrics in early Multiple Sclerosis. Mult Scler Relat Disord. 2021;53:103031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Simpson-Yap S, Oddy WH, Taylor B, et al. ; Ausimmune/AusLong Investigators Group. High Prudent diet factor score predicts lower relapse hazard in early multiple sclerosis. Mult Scler. 2021;27(7):1112–24. [DOI] [PubMed] [Google Scholar]
  • [135].Hadgkiss EJ, Jelinek GA, Weiland TJ, et al. The association of diet with quality of life, disability, and relapse rate in an international sample of people with multiple sclerosis. Nutr Neurosci. 2015;18(3):125–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Fitzgerald KC, Tyry T, Salter A, et al. Diet quality is associated with disability and symptom severity in multiple sclerosis. Neurology. 2018;90(1):e1–e11. [DOI] [PubMed] [Google Scholar]
  • [137].Saul A, Taylor BV, Blizzard L, et al. Associations between diet quality and depression, anxiety, and fatigue in multiple sclerosis. Mult Scler Relat Disord. 2022;63:103910. [DOI] [PubMed] [Google Scholar]
  • [138].Simpson-Yap S, Nag N, Probst Y, et al. Prospective associations of better quality of the diet with improved quality of life over 7.5 years in people with multiple sclerosis. Mult Scler Relat Disord. 2022;60:103710. [DOI] [PubMed] [Google Scholar]
  • [139].Simpson-Yap S, Nag N, Probst Y, et al. Higher-quality diet and non-consumption of meat are associated with less self-determined disability progression in people with multiple sclerosis: a longitudinal cohort study. Eur J Neurol. 2022;29(1):225–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Suliga E, Brola W, Sobas K, et al. Dietary patterns and metabolic disorders in polish adults with multiple sclerosis. Nutrients. 2022;14(9):1927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Abdollahpour I, Jakimovski D, Shivappa N, et al. Dietary inflammatory index and risk of multiple sclerosis: findings from a large population-based incident case-control study. Clin Nutr. 2020;39(11):3402–7. [DOI] [PubMed] [Google Scholar]
  • [142].Keykhaei F, Norouzy S, Froughipour M, et al. Adherence to healthy dietary pattern is associated with lower risk of multiple sclerosis. J Cent Nerv Syst Dis. 2022;14:11795735221092516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Rouzitalab T, Shivappa N, Daneshzad E, et al. Dietary patterns and risk of multiple sclerosis: results of a double-center case-control study in Iran. Nutr Health. 2022:2601060221082379. [DOI] [PubMed] [Google Scholar]
  • [144].Shivappa N, Hebert JR, Behrooz M, et al. Dietary inflammatory index and risk of multiple sclerosis in a case-control study from Iran. Neuroepidemiology. 2016;47(1):26–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Swank RL. Multiple sclerosis; a correlation of its incidence with dietary fat. Am J Med Sci. 1950;220(4):421–30. [PubMed] [Google Scholar]
  • [146].Swank RL. Treatment of multiple sclerosis with low-fat diet. AMA Arch Neurol Psychiatry. 1953;69(1):91–103. [DOI] [PubMed] [Google Scholar]
  • [147].Swank RL, Goodwin J. Review of MS patient survival on a Swank low saturated fat diet. Nutrition. 2003;19(2):161–2. [DOI] [PubMed] [Google Scholar]
  • [148].Swank RL. Treatment of multiple sclerosis with low-fat diet; results of five and one-half years’ experience. AMA Arch Neurol Psychiatry. 1955;73(6):631–44. [DOI] [PubMed] [Google Scholar]
  • [149].Swank RL. Multiple sclerosis: twenty years on low fat diet. Arch Neurol. 1970;23(5):460–74. [DOI] [PubMed] [Google Scholar]
  • [150].Azary S, Schreiner T, Graves J, et al. Contribution of dietary intake to relapse rate in early paediatric multiple sclerosis. J Neurol Neurosurg Psychiatry. 2018;89(1):28–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Wahls TL, Titcomb TJ, Bisht B, et al. Impact of the Swank and Wahls elimination dietary interventions on fatigue and quality of life in relapsing-remitting multiple sclerosis: the WAVES randomized parallel-arm clinical trial. Mult Scler J Exp Transl Clin. 2021;7(3):20552173211035399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Weinstock-Guttman B, Baier M, Park Y, et al. Low fat dietary intervention with omega-3 fatty acid supplementation in multiple sclerosis patients. Prostaglandins Leukot Essent Fatty Acids. 2005;73(5):397–404. [DOI] [PubMed] [Google Scholar]
  • [153].Yadav V, Marracci G, Kim E, et al. Low-fat, plant-based diet in multiple sclerosis: a randomized controlled trial. Mult Scler Relat Disord. 2016;9:80–90. [DOI] [PubMed] [Google Scholar]
  • [154].Ertas Ozturk Y, Merve Helvaci E, Sokulmez Kaya P, et al. Is Mediterranean diet associated with multiple sclerosis related symptoms and fatigue severity? Nutr Neurosci. 2023;26(3):228–34. [DOI] [PubMed] [Google Scholar]
  • [155].Katz Sand I, Benn EKT, Fabian M, et al. Randomized-controlled trial of a modified Mediterranean dietary program for multiple sclerosis: a pilot study. Mult Scler Relat Disord. 2019;36:101403. [DOI] [PubMed] [Google Scholar]
  • [156].Moravejolahkami AR, Paknahad Z, Chitsaz A, et al. Potential of modified Mediterranean diet to improve quality of life and fatigue severity in multiple sclerosis patients: a single-center randomized controlled trial. Int J Food Prop. 2020;23(1):1993–2004. [Google Scholar]
  • [157].Mousavi-Shirazi-Fard Z, Mazloom Z, Izadi S, et al. The effects of modified anti-inflammatory diet on fatigue, quality of life, and inflammatory biomarkers in relapsing-remitting multiple sclerosis patients: a randomized clinical trial. Int J Neurosci. 2021;131(7):657–65. [DOI] [PubMed] [Google Scholar]
  • [158].Razeghi-Jahromi S, Doosti R, Ghorbani Z, et al. A randomized controlled trial investigating the effects of a mediterranean-like diet in patients with multiple sclerosis-associated cognitive impairments and fatigue. Curr J Neurol. 2020;19(3):112–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Riccio P, Rossano R, Larocca M, et al. Anti-inflammatory nutritional intervention in patients with relapsing-remitting and primary-progressive multiple sclerosis: a pilot study. Exp Biol Med (Maywood). 2016;241(6):620–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Jakimovski D, Weinstock-Guttman B, Gandhi S, et al. Dietary and lifestyle factors in multiple sclerosis progression: results from a 5-year longitudinal MRI study. J Neurol. 2019;266(4):866–75. [DOI] [PubMed] [Google Scholar]
  • [161].Bisht B, Darling WG, Grossmann RE, et al. A multimodal intervention for patients with secondary progressive multiple sclerosis: feasibility and effect on fatigue. J Altern Complement Med. 2014;20(5):347–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Bisht B, Darling WG, Shivapour ET, et al. Multimodal intervention improves fatigue and quality of life in subjects with progressive multiple sclerosis: a pilot study. Degener Neurol Neuromuscul Dis. 2015;5:19–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Bisht B, Darling WG, White EC, et al. Effects of a multimodal intervention on gait and balance of subjects with progressive multiple sclerosis: a prospective longitudinal pilot study. Degener Neurol Neuromuscul Dis. 2017;7:79–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Fellows Maxwell K, Wahls T, Browne RW, et al. Lipid profile is associated with decreased fatigue in individuals with progressive multiple sclerosis following a diet-based intervention: results from a pilot study. PLoS One. 2019;14(6):e0218075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Lee JE, Bisht B, Hall MJ, et al. A multimodal, nonpharmacologic intervention improves mood and cognitive function in people with multiple sclerosis. J Am Coll Nutr. 2017;36(3):150–68. [DOI] [PubMed] [Google Scholar]
  • [166].Irish AK, Erickson CM, Wahls TL, et al. Randomized control trial evaluation of a modified Paleolithic dietary intervention in the treatment of relapsing-remitting multiple sclerosis: a pilot study. Degener Neurol Neuromuscul Dis. 2017;7:1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Lee JE, Titcomb TJ, Bisht B, et al. A modified MCT-based ketogenic diet increases plasma beta-hydroxybutyrate but has less effect on fatigue and quality of life in people with multiple sclerosis compared to a modified paleolithic diet: a waitlist-controlled, randomized pilot study. J Am Coll Nutr. 2020;1:13–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Brenton JN, Banwell B, Bergqvist AGC, et al. Pilot study of a ketogenic diet in relapsing-remitting MS. Neurol Neuroimmunol Neuroinflammation. 2019;6(4):e565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Brenton JN, Lehner-Gulotta D, Woolbright E, et al. Phase II study of ketogenic diets in relapsing multiple sclerosis: safety, tolerability and potential clinical benefits. J Neurol Neurosurg Psychiatry. 2022;93(6):637–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Bock M, Karber M, Kuhn H. Ketogenic diets attenuate cyclooxygenase and lipoxygenase gene expression in multiple sclerosis. EBioMedicine. 2018;36:293–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Bock M, Steffen F, Zipp F, et al. Impact of dietary intervention on serum neurofilament light chain in multiple sclerosis. Neurol Neuroimmunol Neuroinflammation. 2022;9(1):e1102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Swidsinski A, Dorffel Y, Loening-Baucke V, et al. Reduced mass and diversity of the colonic microbiome in patients with multiple sclerosis and their improvement with ketogenic diet. Front Microbiol. 2017;8:1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Fitzgerald KC, Bhargava P, Smith MD, et al. Intermittent calorie restriction alters T cell subsets and metabolic markers in people with multiple sclerosis. EBioMedicine. 2022;82:104124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Fitzgerald KC, Vizthum D, Henry-Barron B, et al. Effect of intermittent vs. daily calorie restriction on changes in weight and patient-reported outcomes in people with multiple sclerosis. Mult Scler Relat Disord. 2018;23:33–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Roman SN, Fitzgerald KC, Beier M, et al. Safety and feasibility of various fasting-mimicking diets among people with multiple sclerosis. Mult Scler Relat Disord. 2020;42:102149. [DOI] [PubMed] [Google Scholar]
  • [176].Pommerich UM, Nielsen RO, Overvad K, et al. Diet quality is not associated with late-onset multiple sclerosis risk- A Danish Cohort Study. Mult Scler Relat Disord. 2020;40:101968. [DOI] [PubMed] [Google Scholar]
  • [177].Rotstein DL, Cortese M, Fung TT, et al. Diet quality and risk of multiple sclerosis in two cohorts of US women. Mult Scler. 2019;25(13):1773–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Atuk Kahraman T, Yilmaz M, Yetkin MF, et al. The nutritional status of relapsing-remitting multiple sclerosis (RRMS) patients compared to that of healthy people: a Turkish hospital-based study. Nutr Neurosci. 2022;25(11):2279–87. [DOI] [PubMed] [Google Scholar]
  • [179].Guerrero Aznar MD, Villanueva Guerrero MD, Cordero Ramos J, et al. Efficacy of diet on fatigue, quality of life and disability status in multiple sclerosis patients: rapid review and meta-analysis of randomized controlled trials. BMC Neurol. 2022;22(1):388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Snetselaar LG, Cheek JJ, Fox SS, et al. Efficacy of diet on fatigue and quality of life in multiple sclerosis: a systematic review and network meta-analysis of randomized trials. Neurology. 2023;100(4):e357–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Mannino A, Daly A, Dunlop E, et al. Higher consumption of ultra-processed foods and increased likelihood of central nervous system demyelination in a case-control study of Australian adults. Eur J Clin Nutr. 2023. doi: 10.1038/s41430-023-01271-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Zhang D, Jin W, Wu R, et al. High glucose intake exacerbates autoimmunity through reactive-oxygen-species-mediated TGF-beta cytokine activation. Immunity. 2019;51(4):671–681.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Rezaeizadeh H, Mohammadpour Z, Bitarafan S, et al. Dietary fish intake and the risk of multiple sclerosis: a systematic review and meta-analysis of observational studies. Nutr Neurosci. 2022;25(4):681–9. [DOI] [PubMed] [Google Scholar]
  • [184].Swank RL, Dugan BB. Effect of low saturated fat diet in early and late cases of multiple sclerosis. Lancet. 1990;336(8706):37–9. [DOI] [PubMed] [Google Scholar]
  • [185].Simopoulos AP. Omega-3 fatty acids in inflammation and autoimmune diseases. J Am Coll Nutr. 2002;21(6):495–505. [DOI] [PubMed] [Google Scholar]
  • [186].Simopoulos AP. The importance of the ratio of omega-6/omega-3 essential fatty acids. Biomed Pharmacother. 2002;56(8):365–79. [DOI] [PubMed] [Google Scholar]
  • [187].Salvador R, Zhang A, Horai R, et al. Microbiota as drivers and as therapeutic targets in ocular and tissue specific autoimmunity. Front Cell Dev Biol. 2020;8:606751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Floyd JL, Grant MB. The gut-eye axis: lessons learned from murine models. Ophthalmol Ther. 2020;9(3):499–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Makki K, Deehan EC, Walter J, et al. The impact of dietary fiber on gut microbiota in host health and disease. Cell Host Microbe. 2018;23(6):705–15. [DOI] [PubMed] [Google Scholar]
  • [190].Guan ZW, Yu EZ, Feng Q. Soluble dietary fiber, one of the most important nutrients for the gut microbiota. Molecules. 2021;26(22):68026802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Cronin P, Joyce SA, O’Toole PW, et al. Dietary fibre modulates the gut microbiota. Nutrients. 2021;13(5):1655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Berer K, Martinez I, Walker A, et al. Dietary non-fermentable fiber prevents autoimmune neurological disease by changing gut metabolic and immune status. Sci Rep. 2018;8(1):10431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Fettig NM, Robinson HG, Allanach JR, et al. Inhibition of Th1 activation and differentiation by dietary guar gum ameliorates experimental autoimmune encephalomyelitis. Cell Rep. 2022;40(11):111328. [DOI] [PubMed] [Google Scholar]
  • [194].Kuhle J, Kropshofer H, Haering DA, et al. Blood neurofilament light chain as a biomarker of MS disease activity and treatment response. Neurology. 2019;92(10):e1007–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Disanto G, Barro C, Benkert P, et al. ; Swiss Multiple Sclerosis Cohort Study Group. Serum Neurofilament light: a biomarker of neuronal damage in multiple sclerosis. Ann Neurol. 2017;81(6):857–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Bittner S, Oh J, Havrdova EK, et al. The potential of serum neurofilament as biomarker for multiple sclerosis. Brain. 2021;144(10):2954–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Benkert P, Meier S, Schaedelin S, et al. ; NfL Reference Database in the Swiss Multiple Sclerosis Cohort Study Group. Serum neurofilament light chain for individual prognostication of disease activity in people with multiple sclerosis: a retrospective modelling and validation study. Lancet Neurol. 2022;21(3):246–57. [DOI] [PubMed] [Google Scholar]
  • [198].Cabrera-Mulero A, Tinahones A, Bandera B, et al. Keto microbiota: a powerful contributor to host disease recovery. Rev Endocr Metab Disord. 2019;20(4):415–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Ang QY, Alexander M, Newman JC, et al. Ketogenic diets alter the gut microbiome resulting in decreased intestinal Th17 cells. Cell. 2020;181(6):1263–1275.e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Ye F, Li XJ, Jiang WL, et al. Efficacy of and patient compliance with a ketogenic diet in adults with intractable epilepsy: a meta-analysis. J Clin Neurol. 2015;11(1):26–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Zhu H, Bi D, Zhang Y, et al. Ketogenic diet for human diseases: the underlying mechanisms and potential for clinical implementations. Signal Transduct Target Ther. 2022;7(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Goldberg EL, Shchukina I, Asher JL, et al. Ketogenesis activates metabolically protective gammadelta T cells in visceral adipose tissue. Nat Metab. 2020;2(1):50–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [203].Athinarayanan SJ, Adams RN, Hallberg SJ, et al. Long-term effects of a novel continuous remote care intervention including nutritional ketosis for the management of type 2 diabetes: a 2-year non-randomized clinical trial. Front Endocrinol (Lausanne). 2019;10:348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Lomer MC. Review article: the aetiology, diagnosis, mechanisms and clinical evidence for food intolerance. Aliment Pharmacol Ther. 2015;41(3):262–75. [DOI] [PubMed] [Google Scholar]
  • [205].Reese D, Shivapour ET, Wahls TL, et al. Neuromuscular electrical stimulation and dietary interventions to reduce oxidative stress in a secondary progressive multiple sclerosis patient leads to marked gains in function: a case report. Cases J. 2009;2:7601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Chenard CA, Rubenstein LM, Snetselaar LG, et al. Nutrient composition comparison between a modified paleolithic diet for multiple sclerosis and the recommended healthy U.S.-style eating pattern. Nutrients. 2019;11(3):537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [207].Saresella M, Mendozzi L, Rossi V, et al. Immunological and clinical effect of diet modulation of the gut microbiome in multiple sclerosis patients: a pilot study. Front Immunol. 2017;8:1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Shinto L, Calabrese C, Morris C, et al. A randomized pilot study of naturopathic medicine in multiple sclerosis. J Altern Complement Med. 2008;14(5):489–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Dean C, Parks S, Titcomb TJ, et al. Facilitators of and barriers to adherence to dietary interventions perceived by women with multiple sclerosis and their support persons. Int J MS Care. 2022;24(5):235–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Elkhalii-Wilhelm S, Sippel A, Riemann-Lorenz K, et al. Experiences of persons with Multiple Sclerosis with lifestyle adjustment-A qualitative interview study. PLoS One. 2022;17(5):e0268988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Russell RD, Black LJ, Begley A. Navigating dietary advice for multiple sclerosis. Health Expect. 2021;24(3):853–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Silveira SL, Richardson EV, Motl RW. Desired resources for changing diet among persons with multiple sclerosis: qualitative inquiry informing future dietary interventions. Int J MS Care. 2022;24(4):175–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].AlAmmar WA, Albeesh FH, Ibrahim LM, et al. Effect of omega-3 fatty acids and fish oil supplementation on multiple sclerosis: a systematic review. Nutr Neurosci. 2021;24(7):569–79. [DOI] [PubMed] [Google Scholar]
  • [214].Yadav SK, Boppana S, Ito N, et al. Gut dysbiosis breaks immunological tolerance toward the central nervous system during young adulthood. Proc Natl Acad Sci U S A. 2017;114(44):E9318–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Wekerle H. The gut-brain connection: triggering of brain autoimmune disease by commensal gut bacteria. Rheumatology (Oxford). 2016;55(suppl 2):ii68–75. [DOI] [PubMed] [Google Scholar]
  • [216].Ochoa-Reparaz J, Kirby TO, Kasper LH. The gut microbiome and multiple sclerosis. Cold Spring Harb Perspect Med. 2018;8(6):a029017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Kumar N, Sahoo NK, Mehan S, et al. The importance of gut-brain axis and use of probiotics as a treatment strategy for multiple sclerosis. Mult Scler Relat Disord. 2023;71:104547. [DOI] [PubMed] [Google Scholar]
  • [218].Farshbafnadi M, Agah E, Rezaei N. The second brain: the connection between gut microbiota composition and multiple sclerosis. J Neuroimmunol. 2021;360:577700. [DOI] [PubMed] [Google Scholar]
  • [219].Correale J, Hohlfeld R, Baranzini SE. The role of the gut microbiota in multiple sclerosis. Nat Rev Neurol. 2022;18(9):544–58. [DOI] [PubMed] [Google Scholar]
  • [220].Berer K, Mues M, Koutrolos M, et al. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature. 2011;479(7374):538–41. [DOI] [PubMed] [Google Scholar]
  • [221].Cannas D, Loi E, Serra M, et al. Relevance of essential trace elements in nutrition and drinking water for human health and autoimmune disease risk. Nutrients. 2020;12(7):2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Hesamian MS, Eskandari N. Potential role of trace elements (Al, Cu, Zn, and Se) in multiple sclerosis physiopathology. Neuroimmunomodulation. 2020;27(4):163–77. [DOI] [PubMed] [Google Scholar]
  • [223].Nirooei E, Kashani SMA, Owrangi S, et al. Blood trace element status in multiple sclerosis: a systematic review and meta-analysis. Biol Trace Elem Res. 2022;200(1):13–26. [DOI] [PubMed] [Google Scholar]
  • [224].Sarmadi M, Bidel Z, Najafi F, et al. Copper concentration in multiple sclerosis: a systematic review and meta-analysis. Mult Scler Relat Disord. 2020;45:102426. [DOI] [PubMed] [Google Scholar]
  • [225].Titcomb TJ, Brooks L, Smith KL, et al. Change in micronutrient intake among people with relapsing-remitting multiple sclerosis adapting the swank and wahls diets: an analysis of weighed food records. Nutrients. 2021;13(10):35073507. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Immunometabolism (Cobham (Surrey, England) are provided here courtesy of Wolters Kluwer Health

RESOURCES