1. Introduction
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system (CNS) that affects 2.5 million people worldwide [1], There is currently no cure for MS; however, recent studies demonstrating the influence of diet and the gut microbiota on disease suggest new therapeutic avenues. While nutrition and microorganisms have previously been associated with MS through epidemiological studies, several factors have contributed to the resurgence in attention towards the host-microbe axis. First is the recognition of the importance of the gut microbiota in the development of host immunity. Second is the establishment of gnotobiotic mouse models that allow for more precise investigation of the gut microbiota. Third is a greater appreciation for the role of metabolism, and, by extension, host dietary intake, in host immune function. While difficult to study, the multidirectional relationship between host diet, commensal gut microbiota, and host immunity will be important to understand in order to leverage diet and microbiota-based interventions for the benefit of MS patients.
In this review, we discuss major dietary components and diet regimens that have been associated with MS and the most commonly used preclinical model of MS, experimental autoimmune encephalomyelitis (EAE). We examine recent evidence supporting the role of commensal gut microbiota in the development and progression of disease. We further consider our current understanding of the relationship between host diet and commensal gut microbiota generally before highlighting studies addressing this relationship in the context of EAE and MS. Lastly, we close by posing critical questions related to the interactions between diet, gut microbiota, and MS that must be answered in order to translate diet- and microbiota-based therapies to MS patients.
2. Diet and multiple sclerosis
It is important to precede a review of the literature concerning diet and MS with a note that multiple systematic reviews of dietary intervention trials for MS find no strong evidence for the use of diet in treating MS due to the lack of robust clinical trial design [2,3]. These systematic reviews do not necessarily dismiss the potential benefit of nutritional intervention in MS, but they highlight the challenges of studying dietary interventions in disease. First, many dietary studies use self-reported diet data, which is limited in terms of accuracy. Second, many diet studies are observational/retrospective and do not randomize subjects to diet regimens, thus confounding the results. Third, studies are often limited in population size and duration and thus may miss subtle effects. While the utility of diet-based interventions to manage MS remains tentative, here we focus our review on dietary regimens and specific components that have been studied in MS patients and/or preclinical models of MS with special attention to underlying mechanism, when applicable. The following dietary regimens and components represent promising candidates for more robust follow-up clinical studies.
2.1. Dietary regimens
2.1.1. The Swank diet
The Swank Diet is a low-fat diet developed by Dr. R. L. Swank aimed to benefit patients with MS [4]. The diet was based on an early observation in 1950 that the incidence of MS appeared to correlate with fat consumption [5]. In a follow-up, 34-year-long, non-randomized study of low-fat diets in MS patients, it was found that MS patients that consumed more fat had higher rates of disability compared to MS patients that limited their fat intake; of the patients that began the study with minimum disability, patients that limited their fat intake to below 20 g of fat per day had a lower final mean neurological grade versus patients that consumed 20 g or more of fat per day [6].
2.1.2. Caloric restriction
Calorie restriction regimens that under nourish, but are not mal-nourishing, have been shown to extend the lifespan of model organisms [7], in part by reducing inflammation associated with aging [8,9]. Fasting has been demonstrated to suppress disease in a number of different EAE models [10–12]. In addition, intermittent fasting has also been shown to suppress EAE by suppressing the proinflammatory T helper 17 (Th17) response, inducing anti-inflammatory regulatory T cells (Tregs), and altering the microbiota (discussed in more detail below) [13]. Based on these results, a small, 15-day-long pilot study was performed in which patients with relapse-remitting MS (RRMS) on an intermittent energy restriction diet demonstrated a decrease in circulating lymphocytes, compared to patients who ate ad libitum, and changes were seen in the microbiota at the phylum level that resembled those seen in EAE [13]. More recently, a fasting mimicking diet has been shown to suppress EAE by inducing leukocyte apoptosis, and a 3-month-long trial of the diet improved self-reported quality of life in RRMS patients [14].
2.1.3. Ketogenic diet
A ketogenic diet is generally defined as a low-carbohydrate, high-fat diet that induces the production of ketone bodies as an energy source [15]. A ketogenic diet has been shown to suppress EAE by reducing proinflammatory T helper 1 (Th1) and Th17 responses, reducing reactive oxygen species production, and increasing the proportion of Tregs [14,16]. Pilot studies of the ketogenic diet in RRMS showed improvement in self-reported quality of life, fatigue, and depression over a 3- to 6-month period [14,17], though it will be interesting to perform more longitudinal studies with more objective measures including functional testing and magnetic resonance imaging (MRI).
2.1.4. Western diet and obesity
The rise in obesity in Western countries [18] and prevalence of MS in the Western world [19] has spurred recent investigations into the relationship between the Western diet, obesity, and MS. A Western diet is characterized by excess consumption of refined and processed food, red meat, fat, sugar, and salt as well as reduced consumption of fruits and vegetables [20]. In mice fed a high-fat, high-sugar diet mimicking the Western diet for 18 weeks, their gut microbiota were altered and the concentration of anti-inflammatory short chain fatty acids (SCFAs) were decreased. This supports work in EAE models demonstrating that mice fed a high-fat diet exhibit more severe disease [21]. Furthermore, long chain fatty acids, which are enriched in processed foods typical of the Western diet, have been shown to exacerbate EAE by promoting the differentiation of proinflammatory Th1 and Th17 cells [22]. In a small pilot study, MS patients on a Western diet were shown to have a significant increase in proinflammatory Th17 cells associated with an increase in their expanded disability status scale (EDSS) and a higher relapse rate compared to MS patients on a high-vegetable, low protein diet over a 12 month follow-up period [22].
Obesity, defined by excess body fat and weight for a given height, is intimately tied to the caloric excess associated with the Western diet [18]. In EAE models, obese mice exhibit more severe disease associated with an increase in proinflammatory Th17 cells and a differential gene expression signature in the brain [21,23,24]. This agrees with epidemiological studies that have associated high body mass index (BMI) early in life with a two-fold increased risk of MS [25,26]. This effect, however, was not seen in an Italian case-control population [27]. In pediatric patients with either clinically isolated syndrome (CIS), an initial neurological lesion that may progress to clinical definite MS (CDMS), or CDMS, obesity was associated with increased morbidity [28].
2.1.5. Plant-based diets
Plant-based diets, including vegetarian and vegan diets, have been shown to have anti-inflammatory effects mediated by plant-derived compounds. Such anti-inflammatory compounds include antioxidant polyphenols and carotenoids, as well as fiber, which is metabolized by gut microbiota to produce SCFAs with anti-inflammatory effects. Indeed, in a prospective, randomized trial of healthy male patients, increasing the consumption of carotenoid-rich fruits and vegetables over the course of four weeks resulted in a decrease in the inflammatory biomarker C-reactive protein [29]. In a study of a very low-fat, plant-based diet, MS patients that adhered to the diet for a year versus patients on the control diet had no difference in terms of EDSS, relapse rate, and MRI lesions; however, patients had improvements in BMI, metabolic biomarkers including insulin and cholesterol, and self-reported fatigue [30]. Interestingly, in a pilot study in which healthy omnivores switched to a lacto-ovo vegetarian diet (a plant-based diet that allows the consumption of eggs and dairy) for three months and were compared to long-term omnivores and long-term lacto-ovo vegetarians, there was a decrease in expression of genes related to pro-inflammatory lipopolysaccharide in the gut microbiome of long-term vegetarians that was not seen in either the long-term omnivores nor the short-term vegetarians [31]. These data suggest that while plant-based diets can modulate inflammatory processes, beneficial anti-inflammatory effects may require long-term adherence and warrant further study.
2.1.6. Mediterranean diet
The Mediterranean diet emphasizes fruits, vegetables, whole grains, and unsaturated fats; encourages intake of dairy, fish, and ethanol in moderation; and limits meat consumption [32]. The Mediterranean diet is proposed to influence inflammation by increased consumption of antioxidants including polyphenols [33]. In one prospective study, healthy patients on the Mediterranean diet exhibited a significant reduction in circulating leukocytes and platelets [34], however inflammatory markers C-reactive protein and interleukin (IL)-6 were not changed compared to patients on a control diet. A retrospective case-control study of patients with CIS suggests that adherence to a Mediterranean diet, with consumption of unprocessed red meat, is associated with a lower risk of CIS [35]. This result agreed with another case-control study of MS patients that correlated Mediterranean diet adherence with a reduced risk of MS [36]. However, another case control study of MS patients found no association between a Mediterranean diet and the risk of developing MS [37]. A pilot study randomizing female MS patients onto the Mediterranean diet or a control diet for six months demonstrated a reduction in EDSS and improved fatigue [38].
2.2. Dietary components
2.2.1. Vitamin D
Vitamin D is a fat-soluble vitamin, best characterized for its role in calcium homeostasis that can be taken in via diet or synthesized upon sun exposure [39]. Epidemiological studies of MS patients have shown an increase in MS prevalence in areas farther from the equator, suggesting that sun exposure, and by extension, vitamin D, may be protective against MS [40–42]. Such epidemiological evidence is also supported by genome wide association studies of MS patients that have identified vitamin D related genes (CYP27B1, encodes for 1-alpha-hydroxylase that is required to activate vitamin D; CYP24A1, encodes for 24-hydroxylase that metabolizes active vitamin D to an inactive form) as MS risk alleles (CYP27B1: odds ratio [OR] 1.11, 95% confidence interval [Cl] 1.06–1.15; CYP24A1: OR 1.12, 95% Cl 1.08–1.16) [43], In vivo, Vitamin D has been shown to suppress EAE prophylactically and therapeutically [44–46]. In vitro, vitamin D has been shown to suppress antigen-stimulated proliferation of lymphocytes and limit antibody production [47–49]. Studies investigating the effect of vitamin D on T cell differentiation have shown that vitamin D can suppress pathogenic Th1 [50] and Th17 [51] differentiation and induce protective Tregs [52]. The effects of vitamin D on the adaptive immune system are, in part, due to the tolerogenic effects of vitamin D on dendritic cells [53–55]. Surprisingly, mice that lack the vitamin D receptor or are fed a vitamin D-deficient diet exhibit less severe EAE [56]. In addition, mice that lack either of the hydrolase enzymes that are required to convert vitamin D to its biologically active form exhibit the same severity of EAE as wild-type mice, despite CYP27B1 being identified as a risk allele for MS [56]. This discrepancy underscores the importance of a minimum level of vitamin D signaling required to stimulate an immune response, as vitamin D signaling can potentiate canonical T cell receptor signaling in naïve T cells [57], but leaves open the possibility of a higher, therapeutic dose of vitamin D that may benefit MS patients.
Clinical trials of vitamin D in the context of MS have produced mixed results. There is evidence to suggest that prophylactic vitamin D supplementation can reduce the risk of developing MS: dietary intake of vitamin D during pregnancy was found to reduce the risk of developing MS in offspring (relative risk [RR] 0.57, 95% Cl 0.35–0.91, comparing offspring from mothers in the highest quartile of dietary vitamin D consumption to those in the lowest) [58], and vitamin D levels in patients with CIS are inversely correlated with progression to CDMS (hazard ratio [HR] 0.48, 95% Cl 0.25–0.91, for a 50 nmol/L increase in vitamin D) [59]. Additionally, vitamin D administration may benefit patients with established MS. Vitamin D levels are inversely associated with the appearance of new MS lesions, as detected by MRI (RR 0.53, 95% Cl 0.37–0.78, comparing RRMS patients with vitamin D levels greater than 100 nmol/L to RRMS patients with vitamin D levels between 50.0 and 74.9 nmol/L) [60], and high-dose vitamin D administration (ranging from 10,000 to 40,000 IU/day) was sufficient to decrease the production of proinflammatory IL-17 in peripheral blood CD4+ T cells and suppress proliferation of peripheral blood T cells in MS patients [61,62]. Still, confounding trials suggest that there is no relationship between serum vitamin D levels at birth and development of MS (OR 1.0, 95% Cl 0.68–1.44, comparing the highest quintile of vitamin D to the lowest) [63], nor between high-dose vitamin D (approximately 6000 IU/day to maintain target serum vitamin D concentration of 130–175 nmol/L) and MS progression [64]. A meta-analysis of studies investigating vitamin D in RRMS patients found no association with vitamin D and the risk of relapse (OR 0.98, 95% Cl 0.45–2.16) [65]. Of the specific dietary components associated with MS, vitamin D is among the earliest described and most studied, but more robust studies are needed to investigate and elucidate the potential benefit of vitamin D in MS.
2.2.2. Vitamin A
Like vitamin D, vitamin A is a fat-soluble vitamin with immunomodulatory effects. While vitamin A-deficient mice have blunted regulatory T helper 2 (Th2) responses [66], supplementation with retinoic acid, the active metabolite of vitamin A, potentiates Treg induction [67,68]. Interestingly, retinoic acid also limits the expression of retinoic-acid-receptor-related orphan receptor-γt, the master transcription factor for proinflammatory Th17 cells [67], but is required, at a lower level, for Th17 differentiation [69]. Thus, vitamin A is able to fine tune the immune response based on its local concentration. Work in EAE supports a predominantly beneficial effect of vitamin A and its derivatives to suppress neuroinflammation: 13-cis-retinoic acid, a retinoid related to vitamin A, was found to limit EAE by limiting leukocyte proliferation associated with decreased IL-2 production [70], and all-trans retinoic acid was also found to suppress leukocyte proliferation and skew the immune response to a regulatory Th2 response in EAE [71]. All-trans retinoic acid has also been shown to limit antigen presentation in dendritic cells and was associated with a decrease in proinflammatory Th1 and Th17 polarization in EAE [72]. The pleiotropic effects of vitamin A on both the innate and adaptive arms of the immune system make it a promising candidate for translation to MS patients.
Vitamin A and related retinoids are already used clinically to treat acne and psoriasis [73], however the clinical utility of vitamin A/retinoid supplementation in MS is not clear. MS patients are reported to have lower levels of retinol (mean 2.07 μmol/L, standard deviation 0.21 versus mean 2.53 μmol/L, standard deviation 0.26) and beta carotene (mean 0.41 μmol/L, standard deviation 0.13 versus mean 0.63 μmol/L, standard deviation 0.08), indicators of vitamin A status, compared to matched, healthy controls [74]. This observation also holds true comparing MS patients to patients with noninflammatory neurological disease (mean 46.56 μg/dL, standard deviation 15.45 versus mean 61.50 μg/dL, standard deviation 17.35) [75]. Serum retinol has also been inversely associated with MRI lesions in RRMS patients (OR 0.54, 95% CI 0.32–0.97, for every 1 μmol/L increase in serum retinol) [76]. Promisingly, combining all-trans retinoic acid with interferon β (IFNβ), a disease modifying therapy for MS, has an additive effect in restoring suppressor T cell function in peripheral blood mononuclear cells (PBMCs) from MS patients [77]. However, in a prospective study of RRMS patients, vitamin A was not associated with relapse rates (RR 1.2, 95% Cl 0.8–1.8, comparing RRMS patients with serum levels of all-trans retinoic acid less than 2.9 μmol/L to patients with greater than 3.7 μmol/L) [78]. Given the amount of evidence supporting the immunomodulatory role of vitamin A in preclinical models and the growing literature suggesting a possible benefit of vitamin A supplementation in MS patients, further clinical investigation of vitamin A is warranted.
2.2.3. Other vitamins
While vitamins D and A have been studied extensively in the context of MS, relatively less is known regarding the effect of other vitamins [79,80]. Vitamin B1 (thiamine) is an important cofactor for metabolic enzymes. Deficiency of thiamine in EAE results in exacerbated disease associated with an increase in chemokine production, T cell infiltration, and proinflammatory Th1 and Th17 cells. There is a case report of a small cohort of MS patients treated with high-dose thiamine that observed improvements in self-reported fatigue, however there was no placebo-control in this study [81]. Vitamin B2 (riboflavin) is an antioxidant and important for the process of myelination [82]. Supplementation of riboflavin has a modest effect on suppressing EAE associated with an increase in the expression of brain-derived neurotrophic factor in the CNS [83]. Studies of riboflavin in MS patients are equivocal. A case-control study identified a significant negative linear correlation between the level of riboflavin intake and the risk of developing MS [84]. In contrast, a randomized clinical trial of riboflavin supplementation in MS for six months found no difference in EDSS compared to MS patients given placebo [85]. Vitamin B3 (niacin) is used to make the cofactors nicotinamide adenine dinucleotide and nicotinamide adenine dinucleotide phosphate that are used in metabolic processes, cell signaling, and DNA repair [86]. Supplementation with niacin in EAE results in less severe disease associated with reduced immune cell infiltration into the CNS, increased neurogenesis, and increased proliferation of oligodendrocytes in vitro [87]. However, no clinical trial has been performed to investigate the effect of niacin supplementation in MS patients. Vitamin B6 (pyridoxine) can be interconverted into a number of forms, of which pyridoxal 5’-phosphate is the active form that can act as a cofactor for metabolic reactions [88]. Supplementation with pyridoxamine, another form of vitamin B6, has no effect on the severity of EAE [89] and no trials of vitamin B6 supplementation in MS have been reported. Vitamin B7 (biotin) is an important cofactor for enzymes related to metabolism and cell growth and is proposed to have neuroprotective effects through supporting the metabolism of neurons and glial cells in the CNS [90]. In a progressive EAE model, biotin supplementation had no effect on disease progression [91]. However, an uncontrolled pilot study of high-dose biotin supplementation in secondary progressive MS (SPMS) and primary progressive MS (PPMS) patients demonstrated evidence of clinical improvement as measured by clinical, electrophysiological, and nuclear magnetic resonance spectroscopic assessments [92]. This was followed up with a randomized clinical trial that demonstrated that 12.6% of SPMS or PPMS patients had reduced MS-associated disability after high-dose biotin supplementation at 12 months with significant overall improvement compared to placebo [93]. Vitamins B9 (folate) and B12 (cobalamin) are important cofactors in metabolism and nucleic acid synthesis [94]. Cobalamin is needed to regenerate active folate, thus deficiency in cobalamin can precipitate folate deficiency. Cobalamin supplementation plus IFNβ treatment in EAE was superior to IFNβ treatment alone [95]. In a number of retrospective studies of MS patients, cobalamin was found to be reduced in the serum [96] and cerebrospinal fluid [97]. These reports were supported by a randomized, placebo-controlled trial in which Vitamin B12 and folate supplementation resulted in improvements in self-reported physical and mental quality of life measures in RRMS patients [98]. Of note, certain constituents of the gut microbiota have the ability to synthesize B vitamins [99].
Beyond the B vitamins, vitamin C (ascorbic acid) is an antioxidant that is important for collagen synthesis, wound healing, and the production of neurotransmitters [100]. Therapeutic treatment with ascorbic acid failed to suppress established EAE [101], however L-as-corbyl-2-phosphate, a form of vitamin C, enhanced remyelination in response to cuprizone-induced demyelination [102]. A case-control study reported an inverse correlation with serum vitamin C and MS risk [74]. Vitamin E is an antioxidant that include tocopherols and tocotrienols [103]. Supplementing vitamin E [104] or a vitamin E derivative [105] in EAE results in less severe disease associated with decreased T cell proliferation, muted Th1 response, and reduced demyelination. In chemically-induced demyelination models, vitamin E and a vitamin E derivative have also been shown to enhance remyelination [105,106]. A case-control study of SPMS patients reported decreased serum α-tocopherol in patients compared to controls [74]. This agrees with a prospective cohort study of RRMS patients treated with IFNβ, in which increased serum α-tocopherol was associated with a reduced risk of MRI disease activity [107]. Vitamin K is important for blood coagulation and includes vitamin K1 (phylloquinone), which is derived from plants, and vitamin K2 (menaquinone), which is derived from the metabolism of vitamin K1 by gut microbiota [108]. Prophylactic vitamin K2 supplementation suppressed EAE associated with decreased immune cell infiltration into the CNS and decreased expression of major histocompatibility complex class II and inducible nitric oxide synthase [109]. In contrast, a vitamin K-deficient diet does not affect remyelination in a cuprizone-induced demyelination model [110]. In a case-control study of MS patients, vitamin K2 was found to be decreased compared to controls [111].
2.2.4. Salt
Recent attention has been given to the effect of dietary salt intake on MS given the excessive salt consumption characteristic of the Western diet [112]. Fundamental work investigating the effects of hypertonic saline has shown that the resulting osmotic stress can activate immune cells [113] and stimulate proinflammatory cytokine production [114]. This observation is in agreement with studies in EAE in which a high salt diet induces proinflammatory Th17 cells and exacerbates disease through both a direct [115,116] and gut microbiota-mediated mechanism (discussed in more detail below) [117]. In the direct mechanism, an increase in salt concentration upregulates the expression of serum glucocorticoid kinase 1 (SGK1), which is downstream of p38/mitogen-activated protein kinase [116] and IL-23 signaling [115]. While SGK1 is reported to regulate sodium intake via phosphorylation of epithelial sodium channels, SGK1 can also phosphorylate Foxo1, which deactivates Foxo1, promotes IL-23 signaling, and drives Th17 effector function [115]. Interestingly, a high salt diet induces Th17 differentiation most strongly in the gut lamina propria [115], suggesting that gut-resident immune cells are especially sensitive to the proinflammatory effects of dietary salt.
Clinical investigation into the immunomodulatory effects of a high salt diet are just beginning. Supplementing sodium chloride in healthy patients (additional 6 g of sodium chloride per day above baseline salt intake) for 14 days was sufficient to observe a modest increase in proinflammatory IL-17+ tumor necrosis factor (TNF)+ CD4+ T cells [117]. Indeed, high salt intake (greater than 4.8 g of sodium per day) in MS patients was found to increase disease activity compared to patients with low salt intake (less than 2 g of sodium per day) (exacerbation rate 3.95, 95% Cl 1.4–11.2) [118]. However, other studies in adult patients with CIS (HR of converting to CDMS per 1 g increase in sodium intake 0.91, 95% Cl 0.67–1.24) [119], adult patients with MS (HR of developing MS 0.98, 95% Cl 0.74–1.30, comparing highest and lowest quintiles) [120], and pediatric MS patients (OR of excess sodium intake 1.05, 95% CI 0.67–1.64, comparing pediatric MS cases to controls; HR of relapse 1.37, 95% Cl 0.74–2.51, comparing the high tertile of sodium intake to the low tertile) [121,122] have not shown an association between salt and the development of MS or the progression of disease. Further work, with an emphasis on randomized clinical trials, will be needed to resolve conflicting data regarding the influence of salt on MS.
2.2.5. Polyphenols
Dietary polyphenols are derived from fruits, vegetables, cereals, legumes, herbs, spices, wine, fruit juices, tea, and coffee [4]. This family of compounds can be structurally categorized into flavonoids, such as quercetin, and non-flavonoids, such as resveratrol. Polyphenols have been shown to influence the immune response through direct antioxidant effects [123] and by suppressing the activity of the pro-inflammatory nuclear transcription factor κB (NF-κB) [124]. To date there has been no study of the effect of polyphenol supplementation in MS patients and results in preclinical MS models have been mixed. Green tea polyphenol epigalloatechin 3 gallate was sufficient to ameliorate EAE and was associated with lower pro-inflammatory cytokine production and reduced T cell proliferation to myelin peptide [125]. Natural flavonoids, including apigenin, hesperidin, luteolin, and quercetin, have also been shown to protect against EAE by modulating the immune response [126–128]. Resveratrol has been reported to support remyelination in a chemically-induced preclinical model of MS [129]. However, resveratrol has also been shown to exacerbate disease in EAE and virus-induced preclinical MS models [130]. In addition, quercetin was also found to mildly ameliorate EAE in one study [128] and exacerbate EAE in another [131].
2.2.6. Thiolic compounds
Thiolic compounds contain a thiol (-SH) functional group and include α-lipoic acid, glutathione, and N-acetylcysteine [4]. In EAE, α-lipoic acid has been shown to suppress disease by limiting T cell infiltration into the CNS [132,133]. In a double-blind, randomized, placebo-controlled trial, lipoic acid supplementation over two years resulted in a reduction in percent change brain volume in SPMS patients, but no significant change in EDSS and a trending increase in T2 lesion volume [134]. Glutathione is a major antioxidant in the CNS [135] and level of glutathione is decreased in the spinal cord of EAE-induced mice [136]. N-acetylcysteine administration has been shown to suppress EAE by increasing glutathione production as well as acting as a reactive oxygen species scavenger itself [137,138]. Oral glutathione supplementation has low bioavailability [139] and limited penetration of the blood-brain barrier [135]. While no trial of glutathione in MS patients has been performed, there is a randomized prospective trial of N-acetylcysteine currently under way (NCT0303601).
2.2.7. Fibers
Fibers, such as inulin and galacto-oligosaccharides, are composed of resistant starch and oligosaccharides that are not digested by the human host, but instead metabolized by colonic bacteria [140]. Fruits, vegetables, whole grains, and legumes are rich in fiber whereas the typical Western diet is low in fiber [141]. As a substrate for bacteria, the abundance of fiber can alter the gut microbiota and is suggested to promote the growth of tolerogenic commensal bacteria. Fibers can also be fermented to SCFAs by bacteria, which themselves have anti-inflammatory properties [142]. Indeed, fiber supplementation has been shown to ameliorate EAE and is associated with an increase in intestinal SCFAs and an altered gut microbiota [143,144]. While no study has been conducted to examine the effects of dietary fiber supplementation specifically in MS patients, there is evidence to suggest that the consumption of fiber-rich foods can benefit MS patients (see plant-based diets).
2.2.8. Short chain fatty acids (SCFAs)
SCFAs include acetic acid, butyric acid, propionic acid, and pentanoic acid and are largely derived from the fermentation of dietary fiber by commensal gut microbiota [145]. SCFAs have been shown to promote tolerogenic responses, via signaling through G-protein coupled receptors and inhibition of histone deacetylases that can inactivate NF-κB, and suppress the expression of proinflammatory cytokines as well as induce anti-inflammatory macrophages, dendritic cells, and regulatory T cells [146–151]. SCFAs also have been shown to affect the maturation and function of microglia, the resident myeloid cell population in the CNS, and support the integrity of the blood-brain barrier [152,153]. The SCFA propionic acid has been shown to ameliorate EAE through the induction of Tregs and a decrease in proinflammatory IL-17 [22]. Additionally, pentanoic acid, butyric acid, and acetic acid have also been shown to ameliorate EAE [144,154] and butyric acid, in particular, has been shown to ameliorate chemically-induced demyelination [155]. A case-control study of SPMS patients found decreased blood concentrations of acetic acid, butyric acid, and propionic acid, but pentanoic acid was unchanged compared to healthy controls [156]. A pre-print suggests that supplementation of propionic acid in healthy volunteers and MS patients can increase Tregs and decrease Th17 with reduced relapse rate and disease progression after one year of supplementation [157].
2.2.9. Long chain polyunsaturated fatty acids (LC-PUFAs)
LC-PUFAs are defined by having more than one double bond in their structure and include omega-3 (n-3) and omega-6 (n-6) fatty acids [158]. Nuts, seeds, fish oil, and fish such as salmon, tuna, mackerel, herring, and sardines are major sources of n-3 LC-PUFAs [159]. In contrast, n-6 LC-PUFAs are derived predominantly from vegetable oils such as soybean and corn oil [160]. Linoleic acid, a major dietary n-6 LC-PUFA, can be converted to arachidonic acid and further metabolized to eicosanoids such as prostaglandins and leukotrienes that have pleotropic effects on the production of pro- and anti-inflammatory cytokines, reactive oxygen species, immunoglobulins, and the proliferation of lymphocytes [161,162]. α-linolenic acid, a major dietary n-3 LC-PUFA, can be converted to eiosapentaeoic acid (EPA) and docosahexanoic acid (DHA) that compete with the same enzymes that metabolize arachidonic acid in order to form alternative eicosanoids that have less biological activity than eicosanoids derived from arachidonic acid [163]. Thus, the Western diet is thought to promote inflammation, in part due to a relative increase in the ratio of n-6 to n-3 LC-PUFAs [164]. In a randomized, placebo-controlled trial, MS patients that were supplemented with high-dose n-3 LC-PUFAs and vitamin D had improvements in EDSS and inflammatory biomarkers over a 12-week period, though their experimental design does not isolate the effect of n-3 LC-PUFAs alone [165]. In a retrospective case-control study, it was found that high fish consumption reduced the risk of developing MS or CIS (OR 0.55, 95% Cl 0.4–0.75) independent of serum vitamin D concentration, and single nucleotide polymorphisms in fatty acid desaturase 2, an enzyme involved in fatty acid biosynthesis, were also found to decrease MS risk [166]. However, in a randomized, double-blind, placebo-controlled trial, MS patients supplemented with n-3 LC-PUFAs EPA and DHA for six months exhibited no difference in relapse rate, quality of life, nor MRI lesion activity [167].
3. Gut microbiota and multiple sclerosis
Closely tied to diet is the role of the commensal gut microbiota in MS. This is unique from the role of infection in MS, which has been associated with an increased risk of developing MS and exacerbation of MS symptoms [168–172]. In contrast to these more transient host-pathogen interactions, host-commensal interactions have effects on physiology throughout the host’s lifespan. In the context of demyelinating disease, this is most evident in germ-free mice that lack an endogenous microbiota and develop less severe EAE compared to conventionally colonized animals [173,174]. This effect can be recapitulated in conventionally colonized mice treated with antibiotics [175,176]. Indeed, specific commensal microorganisms, such as segmented filamentous bacteria [173], have been shown to exacerbate EAE, whereas other commensals such as Bacteroides fragilis [177], Lactobacillus species [117,178–182], and Prevotella histolitica [183] have been shown to ameliorate disease. These results in EAE have been supported by studies in MS patients demonstrating that patients with MS have distinct bacterial taxa that are differentially abundant compared to healthy controls [184–186]. Subsequent studies have demonstrated that colonization of germ-free mice with microbiota derived from MS patients results in more severe EAE than germ-free mice colonized with microbiota from healthy controls [187,188], suggesting that the gut microbiota plays a functional role in disease.
3.1. Identifying an MS-associated microbiota
There is a growing body of literature investigating gut microbial communities in MS patients. While the broad gut bacterial community composition of MS patients is not different from healthy controls, there are strains of bacteria that have been reproducibly found to be altered in MS patients. Among the first studies to examine the gut microbiota in MS, Yamamura and colleagues performed 16S ribosomal RNA (rRNA) sequencing on fecal samples from 20 Japanese RRMS patients and 40 Japanese healthy patients and found that the gut microbiota of MS patients was less similar than the gut microbiota of healthy patients compared to the inter-individual variation in gut microbiota between healthy patients [184]. However, the inter-individual variation in gut microbiota between MS patients was greater than the difference in gut microbiota between MS patients and healthy patients, which may be due to the relatively small cohort size in the MS population and the variability in disease modifying therapies being used in the MS cohort. Still, at the genus level, Faecalibacterium, Prevotella, and Anerostipes were found to be significantly decreased in MS patients compared to healthy patients.
Mangalam and colleagues published a subsequent study examining the gut microbiome of 31 American RRMS patients and 36 age- and sex-matched healthy controls [185]. Gut community analysis revealed that RRMS patients had a distinct gut microbiota from healthy controls, and of note, the gut microbial community of RRMS patients in remission was more similar to healthy controls than the gut microbiota of RRMS patients with active disease. At the genus level, Adlercrutzia, Collinsella, Parabacteroides, Coprobacillus, Lactobacillus, and Haemophilus were found to be significantly decreased in MS patients. Conversely, Flavo-bacterium, Pedobacter, Blautia, Dorea, Mycoplana, and Pseudomonas were found to be significantly increased in MS patients.
In a multi-tiered approach, Weiner and colleagues combined 16S rRNA sequencing with blood and breath analysis of RRMS and healthy controls [186]. For 16S rRNA sequencing, fecal samples were collected from 60 American RRMS patients and 43 American healthy controls and sequenced on two different sequencing platforms that differed in sequencing read length and sequencing depth. While no significant difference was found between broad gut microbial community structure between RRMS patients and healthy controls, Butyricimonas, Paraprevotella, and Slackia were genera identified to be decreased in RRMS patients regardless of sequencing platform. In contrast, Methanobrevibacter and Akkermansia were significantly upregulated in RRMS patients. Interestingly, Haemophilus was found to be both significantly upregulated or downregulated in RRMS patients depending on the sequencing platform used, however Haemophilus was consistently increased when comparing only untreated RRMS patients to healthy controls. Collinsella was not significantly changed between RRMS patients and healthy controls, though it was decreased when comparing only untreated RRMS patients and healthy controls. Cloacibacillus, Prevotella, and Sutterella are significantly decreased in untreated RRMS patients compared to healthy controls, whereas treated RRMS patients have significantly higher relative abundance of these genera. The opposite trend is observed for Megasphaera. Sarcina is significantly decreased only in treated MS patients compared to untreated MS patients. Additionally, Holdemania, Desulfovibrio, Barnesiella, Megamonas, Guggenheimella, Buttiauxella, and Sporanaerobacter were also identified as being differentially abundant between study groups, though significance of these differences and even detection of these genera was dependent on the sequencing platform. Transcriptomic analysis of blood CD4+ T cells and monocytes showed that the abundance of Methanobrevibacter and Akkermansia had a positive correlation with the expression of proinflammatory genes in line with their enrichment in RRMS patients. In contrast, the abundance of Butyricimonas had a negative correlation with proinflammatory genes parsimonious with its depletion in RRMS patients. Additionally, breath methane was measured in a second, independent cohort of RRMS patients and healthy controls as Methanobrevibacter is the dominant methane-producing commensal bacteria in the gut. Breath methane was demonstrably higher in RRMS patients, providing in vivo support for one of their metagenomic results.
Wekerle and colleagues sought to better isolate the effect of the gut microbiota and minimize the effect of host genetic variability by examining the gut microbiota of German monozygotic twin pairs in which one twin had MS (ranging from CIS, RRMS, SPMS, and PPMS) and the other twin was unaffected [188]. Microbiomes were characterized using both 16S rRNA sequencing and metagenomics shotgun sequencing and found no significant differences in overall microbial community structure. Interestingly, when MS patients were subdivided into treated and untreated groups, Akkermansia was significantly increased in untreated MS twins. To functionally test the contribution of the gut microbiota in disease, the fecal microbial communities from five RRMS discordant monozygotic twin pairs were transferred to germ-free transgenic mice that develop spontaneous relapse-remitting EAE. While there was a significant reduction in number of bacterial taxa retrieved from colonized mouse feces compared to their human donor, suggesting an inability of certain human gut commensals to colonize the mouse, mice that received microbiota from the MS-affected twin had a higher incidence of spontaneous EAE compared to mice that received microbiota from the unaffected twin. The genus Ruminococcus was found to be enriched in mice colonized with microbiota from the MS-affected twin while the genera Adlercreuzia, Tannerella, and Sutterella were found to be decreased. Functionally, splenic T cells derived from MS microbiota-colonized mice as well as blood CD4+ T cells of MS-affected twins produced less IL-10 when stimulated.
In a larger scale study, Baranzini and colleagues performed 16S rRNA sequencing on fecal samples from 71 American untreated RRMS patients and 71 American healthy patients [187]. While there were no significant differences in microbial community structure, there were significant differences at a number of taxonomic levels. At the genus level, Prevotella, Serratia, Bacillus, Acidaminococcus, Clostridium, Aquamonas, Parabacteroides, and Phascolarctobacterium were found to be significantly decreased in RRMS patients whereas Acinetobacter, Varibaculum, Corynebacterium, Bifidobacterium, Megamonas, Megasphaera, Klebsiella, ph2, Actinomyces, Mogibacterium, Akkermansia, SMB53, and Bulleidia were found to be significantly increased. At the operational taxonomic unit (OTU) level, in which sequencing reads are clustered based on sequence similarity and assigned an OTU that refers to a bacterial sequence reference database [189], an additional 247 OTUs were differentially abundant in RRMS patients. A number of these OTUs refer to genera identified in their previous analysis (e.g. Acinetobacter, Akkermansia, Parabacteroides, etc.) and others refer to new genera (e.g. Lactobacillus, Suttera, etc.) not identified in their previous analysis. Based on their metagenomic analysis, the strains Acinetobacter calcoeticus, Akkermansia muciniphila, and Parabacteroides distasonis were further validated in in vitro and in vivo systems to test their immunomodulatory properties. A. calcoeticus was found to inhibit the differentiation of Tregs and both A. calcoeticus and A. muciniphila were found to induce the differentiation of Th1 cells in healthy human PBMCs, consistent with their enrichment in RRMS patients. In contrast, P. distasonis was found to induce anti-inflammatory, IL-10-producing Tregs in healthy human PBMCs, in line with its depletion in RRMS patients. These results were largely recapitulated by monocolonization of germ-free or antibiotic-treated mice with these strains, however A. mucinophila was unable to induce Th1 cells in the mouse, possibly due to differences in mouse and human interaction with A. mucinophila. To more broadly investigate the role of the gut microbiota in MS, three fecal donor pairs, each consisting of one RRMS patients and a household control, were randomly chosen to transfer fecal microbiota to germ-free mice. While overall gut microbial community structure was altered in recipient mice compared to their corresponding human donors, suggesting a difference in the ability of bacteria to colonize the mouse gut versus the human gut, the mice that received MS-associated microbiota had distinct community structure from mice that received control-associated microbiota. Furthermore, mice that received MS-associated microbiota exhibited more severe EAE associated with reduced proportions of Tregs.
These studies of the MS-associated microbiome have failed to find a distinct change in broad gut community structure, however changes in certain bacterial taxa have been reproduced. Table 1 lists the various microbial genera identified in gut microbiota studies of MS patients with potential mechanistic links, where available. Table 2 provides the demographics of the patient populations studied in the context of MS-associated gut microbiota. Variability in the results of these and other studies of the MS-associated microbiome raise important methodological and conceptual considerations. In terms of study design, the recruitment of patients from multiple study sites is important for the external validity of the study as it has previously been shown that the microbiota can vary by geographic region [190]. In addition, special considerations should be made when enrolling untreated patients versus patients on various disease modifying therapies (DMTs) as DMTs also have varying effects on the gut microbiota [191]. The protocol for DNA extraction, sequencing, and metagenomic analysis also affects results and adds to the difficulty of comparing studies using different pipelines [192,193]. Inconsistencies apparent in the MS gut microbiota literature may also reflect the inherent difficulty in resolving species-and strain-level taxa through 16S rRNA sequencing, leading to different OTUs defined as the same genera being both increased and decreased in MS patients even within the same study (e.g. Prevotella in Table 1). Fundamentally, these types of case-control metagenomic studies are correlative and act as a starting point for further investigation into mechanistic understanding of the influence of the microbiota on MS. Critically, while differentially abundant microbial strains provide promising targets for investigation, the contribution of non-differentially abundant strains, which may function differently in MS patients compared to healthy controls, should also be considered.
Table 1.
Genera | Abundance MS: controls | Reference(s) in which difference identified | Reference(s) in which opposite difference identified | Mechanistic understanding |
---|---|---|---|---|
Acidaminococcus | increased | [217] | [187] | |
Acinetobacter | increased | [187] |
A. calcoaceticus reduced CD25+ FoxP3+ Tregs in human PBMCs [187] A. calcoaceticus increased differentiation of human peripheral blood CD4+ T cells to IFNγ-producing Th1 cells [187] Antibody cross-reactivity between Acinetobacter and myelin peptides [218] |
|
Actinomyces | increased | [187] | ||
Adlercreutzia | decreased | [185] | ||
Akkermansia | increased | [217] [186] [187] [188] |
A. muciniphila increased differentiation of human peripheral blood CD4+ T cells to IFNγ-producing Th1 cells [187] | |
Anaerococcus | increased | [187] | ||
Anaerostipes | increased | [217] | [184] | |
Aquamonas | decreased | [187] | ||
Atopobium | increased | [217] | ||
Bacillus | decreased | [187] | ||
Bacteroides | decreased | [217] [184] [187] |
[217] | Decreased EAE severity via B. fragilis polysaccharide A-mediated induction of regulatory dendritic cells and Tregs and suppression of Th17s [177,219] B. fragilis expands of regulatory CD39+CD4+ T cells [220] |
Barnesiella | decreased | [186] | ||
Bifidobacterium | increased | [217] [187] |
[187] | Supplementation of Bifidobacterium ameliorates EAE in Lewis rats [221] Supplementation of Bifidobacterium ameliorates EAE in C57BL/6 mice [182] |
Bilophila | increased | [217] | [187] | |
Blautia | increased | [217] [185] [187] |
[217] [187] |
|
Bulleidia | increased | [187] | ||
Buttiauxella | decreased | [186] | ||
Butyricimonas | decreased | [217] [186] [187] |
||
Catenibacterium | increased | [217] | [217] | |
Citrobacter | increased | [217] [187] |
C. rodentium induces inflammatory Th17 cells [222] | |
Cloacibacillus | increased | [186] | ||
Clostridium | decreased | [184] [187] |
[187] |
C. butyricum suppresses EAE associated with decreased Th17 and increased Treg [223] Clostridia consortia induces colonic Treg [224] |
Collinsella | decreased | [185] [186] [187] |
||
Coprobacillus | decreased | [185] [187] |
||
Coprococcus | decreased | [217] [184] [187] |
[217] [187] |
|
Corynebacterium | increased | [187] | ||
Dehalobacterium | increased | [217] | ||
Desulfotomaculum | decreased | [184] | ||
Desulfovibrio | increased | [217] [184] [186] [187] |
[187] | |
Dialister | increased | [217] [187] |
[217] | |
Dorea | decreased | [217] [187] |
[185] | |
Eggerthella | increased | [184] | ||
Enterobacter | increased | [187] | E. lenta does not affect human PBMC differentiation towards Th1 or Treg [187] | |
Eubacterium | increased | [217] [187] |
[184] [187] |
|
Faecalibacterium | decreased | [217] [187] |
[187] | F. prausnitzii decreases Th17 differentiation and increases Treg differentiation in mouse CD4 + T cells va production of butyrate [225] |
Flavobacterium | increased | [185] | ||
Fusobacterium | decreased | [187] | ||
Guggenheimella | decreased | [186] | ||
Haemophilus | decreased | [185] [186] |
[186] | |
Klebsiella | increased | [187] | Intratracheal infection with K. pneumoniae induces Th17 cells [226] | |
Lachnospira | decreased | [217] [184] [187] |
||
Lactobacillus | decreased | [217] [185] [187] |
[217] |
L. reuteri ameliorates EAE with reduced Th1 and Th17 cells [178] L. paracasei DSM 13434, L. plantarum DSM 15312, and L. plantarum DSM 15313 ameliorate EAE via IL-10 producing Tregs and suppression of IFNy and IL-17 production [179] L. helviticus SBT2171 attenuates EAE by reducing IL-6 production, T cell infiltration into the spinal cord, and Th17 proportions [227] L. murinis ameliorates EAE by reducing Th17 cells through indole metabolites [117] L. paracasei ATCC27092 ameliorates EAE [181] L. plantarum A7 ameliorates EAE via induction of Tregs [182] |
Megamonas | increased | [217] [186] [187] |
[184] | |
Megasphaera | increased | [217] [186] [187] |
||
Methanobrevibacter | increased | [217] [186] |
||
Mitsuokella | decreased | [217] | ||
Mogibacterium | increased | [187] | ||
Mycoplana | increased | [185] | ||
Odoribacter | increased | [187] | [187] | |
Oscillospira | decreased | [217] [187] |
[217] | |
Oxalobacter | decreased | [187] | ||
Parabacteroides | decreased | [185] [187] |
[217] | P. distasonis increased differentiation of human peripheral blood CD4+ T cells to CD25+ IL-10+ T cells [187] |
Paraprevotella | decreased | [217] [186] [187] |
[217] | |
Pedobacter | increased | [185] | ||
Peptostreptococcus | decreased | [217] | ||
ph2 | increased | [187] | ||
Phascolarctobacterium | increased | [217] | [187] | |
Porphyromonas | increased | [217] [187] |
Subcutaneous application of P. gingivalis A7436 five days after EAE induction exacerbates disease associated with increased nitric oxide and prostaglandin E2 production from glial cells [228] | |
Prevotella | decreased | [217] [184] [186] [187] |
[217] [187] |
Human gut-derived P. histolitica induced tolerogenic dendritic cells and Tregs and suppressed Th1 and Th1 7 response [183,229] |
Providencia | increased | [217] | ||
Pseudomonas | increased | [185] [187] |
[187] | |
rc4–4 | increased | [217] [187] |
||
Ruminococcus | increased | [217] [187] |
[217] [187] |
R. gnavus produces a polysaccharide that induces TNFα production [230] |
Sarcina | decreased | [186] | ||
Serratia | decreased | [187] | ||
Slackia | decreased | [186] | ||
SMB53 | increased | [187] | ||
Sporanaerobacter | decreased | [186] | ||
Streptococcus | increased | [217] [184] |
[187] | S. pneumoniae infection seven days after EAE induction increases severity of EAE via a Toll-like receptor 2-dependent mechanism with increased serum TNFα and IL-6 and increased expression of MHC-II, CD80, and CD86 in splenic dendritic cells [231] |
Succiniclastum | increased | [217] | ||
Sutterella | decreased | [217] [184] [186] [187] |
[217] | |
Trabulsiella | decreased | [217] | ||
Varibaculum | increased | [187] | ||
Veillonela | increased | [187] | ||
WAL_855D | increased | [217] [187] |
Table 2.
Reference | Demographics |
---|---|
Miyake et al. [184] | 20 Japanese RRMS cases vs 40 healthy controls |
Tremlett et al. [217] | 18 American pediatric MS cases vs 17 healthy age- and sex-matched controls |
Chen et al. [185] | 31 American RRMS cases vs 36 age- and sex-matched controls |
Jangi et al. [186] | 60 American RRMS cases vs 43 healthy controls and 28 untreated American RRMS cases vs 43 healthy controls |
Berer et al. [188] | 17 untreated American discordant twin MS cases vs 34 healthy discordant twin controls |
Cekanaviciute et al. [187] | 71 American RRMS cases vs 71 non-autoimmune controls |
3.2. Clinical trials of microbiota-based interventions
Clinical trials have been and are currently being performed to investigate the potential benefit of probiotic administration in MS patients. In a small, randomized, placebo-controlled trial, MS patients were given either a probiotic supplement containing Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus fermentum, and Bifidobacterium bifidum or placebo for 12 weeks [194]. PBMCs from probiotic-supplemented MS patients had decreased expression of proinflammatory cytokines IL-8 and TNFα, but no difference was observed in the expression of proinflammatory cytokine IL-1. A follow up study by the same group demonstrated improvement in EDSS, self-reported quality of life measures, and biomarkers of inflammation with probiotic supplementation [195]. Another pilot study of VSL3, a probiotic cocktail of Lactobacillus, Bifidobacterium, and Streptococcus species, in MS patients demonstrated that probiotic administration can transiently alter the gut microbiota in MS patients and decrease antigen presentation in peripheral dendritic cells [196]. Further studies are needed to determine whether probiotic regimens can benefit MS progression with long term administration.
3.3. How diet influences the gut microbiota
While it has been appreciated that diet is able to shape gut commensal microorganisms [197,198], our understanding of the interactions that drive diet-based modulation of gut commensal organisms is still expanding [199,200].
Diet can directly affect the gut microbiota by providing substrates to support the growth of gut community members. This is predominantly in the form of indigestible carbohydrates such as resistant starch, inulin, and cellulose that can be degraded by bacteria expressing carbohydrate-active enzymes. Sugars released by such bacteria can further feed subsequent bacteria forming a complex metabolic network whose metabolites include SCFAs, hydrogen gas, lactate, and succinate [201]. Diet can also directly affect the gut microbiota by inhibiting the growth of gut community members as seen with salt [117] and plant-derived compounds including tannins, terpinoids, alkaloids, and flavonoids [202]. In addition, diet can indirectly influence the gut microbiota by influencing host immunity, intestinal barrier function, and production of host-derived secondary products. Intraepithelial lymphocytes in the gut rely on signaling through the aryl hydrocarbon receptor (AhR), the ligands of which can be plant-derived, in order to be maintained, and without AhR ligands bacterial outgrowth of the phylum Bacteroidetes was observed in mice [203]. Vitamin D signaling appears to be critical for the development of tolerogenic dendritic cells in the lamina propria as mice that cannot product the active form of vitamin D have fewer dendritic cells and are associated with increases in Desulfoibrionceae, Bacteroidaceae, and Bacteroidales and decreases in Ruminococcaceae, Lachnospiraceae, Lactobacillceae, and Streptococcaceae [204]. Supplementation of n-3 LC-PUFAs increases intestinal alkaline phosphatase, which can directly act on gut microbes to shift composition by decreasing Escherichia coli and increasing Bifidobacterium [205]. Vitamin D signaling is also critical for the expression of antimicrobial defensins by Paneth cells that can directly inhibit bacteria [206]. Vitamin A deficiency results in increased bile acid that can directly inhibit the growth of bacteria such as Bacteroides vulgatus [207]. Even dietary emulsifiers, which are common additives in processed food, have been shown in mice to disrupt the protective mucus coating of the intestine and alter the mucosa-associated bacterial consortia [208].
The change in gut microbiota occurs rapidly in response to diet, as four days on an animal-based diet in healthy human subjects is sufficient to increase β diversity, enrich bile-resistant bacteria including Bilophila, Alistipes, and Bacteroides, and deplete bacteria able to metabolize plant polysaccharides including Roseburia, Eubacterium, and Ruminococcus [209]. However, the gut microbiota is relatively resilient to dietary perturbation, as returning to a baseline diet after four days on an animal-based diet resulted in a return of β diversity back down to baseline levels [209]. In addition, six weeks of psyllium supplementation in children with irritable bowel syndrome did not result in significant changes in the gut microbiota [210]. Furthermore, two weeks of either white bread or sourdough bread supplementation in healthy human subjects did not result in significant changes in overall community structure compared to each individual’s baseline, however, Anerostipes and Eubacterium were enriched in subjects supplemented with white bread versus sourdough bread [211]. Alterations in gut microbiota have been observed within 24 h of starting a high-fiber/low-fat or low-fiber/high-fat diet [212]. Specific diet-microbiota interactions that have been reported, with special consideration towards diet regimens and dietary components associated with MS, are listed in Table 3. There is notable variation in the response of the gut microbiota to host diet and a relative lack of understanding regarding the mechanisms by which diet alters the gut microbiota. Still, associations between Table 1 and Table 3 suggest relevant crosstalk between diet and gut microbiota in MS. For example, Bacteroides, which has been reproducibly shown to be decreased in MS patients (Table 1), is also enriched with vitamin D in two separate reports (Table 3). Thus vitamin D, beyond its direct immunomodulatory effects, may also act through Bacteroides and other vitamin D-sensitive microbial strains to decrease the risk of MS. Future studies of dietary intervention in MS would benefit greatly by including changes in the gut microbiota as relevant read-out to parse out direct diet-mediated effects versus microbiota-dependent effects.
Table 3.
Diet and associated effects on the gut microbiota | Genera enriched | Genera depleted |
---|---|---|
Caloric restriction | Bacteroides [232] Blautia [13] Faecalibacterium [13] Lactobacillus [232] |
Bifidobacterium [232] Clostridium [232,233] Eubacterium [233] |
Ketogenic diet | Bacteroides [234–236] Desulfovibrio [237] Dorea [234] Escherichia [238] Prevotella [236] |
Acinetobacter [235] Alistipes [236] Barnesiella [236] Bifidobacterium [238] Coprobacter [235] Cronobacter [236] Dialister [238] Enterococcus [236] Erysipelatoclostridium [236] Eubacterium [238] Faecalibacterium [234,235] Lachnospiraceae incertae sedis [235] Leucobacter [235] Pseudomonas [235] Ruminiclostridium [236] Streptococcus [236] |
Western diet | Akkermansia [239] | Butyrivibrio [240,242] |
Bacteroides [240,241] Escherichia [242] Faecalibacterium [240] Shigella [242] |
Prevotella [241,242] Treponema [242] Xylanibacter [242] |
|
Obesity | Akkermansia [243] Alistipes [243] Bacteroides [243,244] Clostridium [243,245,246] Desulfovibrio [243] Eggerthella [243] Escherichia [243] Parabacteroides [243] Ruminococcus [245] Streptococcus [246] Subdoligranulum [243] |
Bacteroides [245,246] Eubacterium [243] Faecalibacterium [243,246] Haemophilus [243] Lachnospira [244] Oscillospira [244] Prevotella [246] Roseburia [243,246] Ruminococcus [246] Sporobacter [245] |
Plant-based diet | Actinobacillus [31,209] Actinoplanes [31] Aggregatibacter [31] Atopobium [31] Bacteroides [247] Blautia [31] Capnocytophaga [31] Clostridium [209,247,248] Cryptobacterium [31] Faecalibacterium [247] Haemophilus [31] Holdemania [248] Klebsiella [249] Lachnospira [250] Micrococcus [31] Neisseria [31] Porphyromonas [31] Prevotella [249,250] Roseburia [248,250] Staphylococcus [31] Subdoligranulum [247 Veillonella [31] |
Aceto bacterium [31] Akkermansia [249] Alistipes [249] Bacteroides [31,249,251] Bifidobacterium [247,251] Bilophila [31,209] Blautia [209] Bulleidia [31] Caldanaerobacter [31] Campylobacter [31] Clostridium [31,209] Collinsella [247,248] Dehalogenimonas [31] Desulfitobaeterium [31] Desulfovibrio [31] Dialister [31] Dorea [31] Eggerthella [31] Escherichia [251] Eubacterium [248] Holdemania [31] Methanosphaera [31] Oscillospira [209] Oxalobacter [31] Parabacteroides [249] Phascolarctobacterium [31] Prevotella [31] Ralstonia [31] Ruegeria [31] Ruminococcus [209] Streptobacillus [31] Streptococcus [247] Syntrophobacter [31] Taylorella [31] Thermoanaerobacter [31] |
Animal-based diet | Alistipes [209] Bacteroides [209] Bilophila [209] Roseburia [252] |
Butyricimonas [252] Eubacterium [209] Parabacteroides [252] Phaseolaretobaeterium [252] Roseburia [209] Ruminoeoeeus [209] |
Mediterranean diet | Bacteroides [246] Catenibacterium [252] Citrobacter [250] Enterorhabdus [253] Eubacterium [254] Faecalibacterium [246] Lachnoclostridium [253] Lachnospira [250] Oseillospira [255] Parabacteroides [246,255] Prevotella [246,250,254] Roseburia [246,255] Ruminococcus [246] |
Clostridium [252] Collinsella [250] Coprobaeillus [250] Enterococcus [250] Escherichia [256] Parabacteroides [253] Prevotella [255] |
Vitamin A | Butyricimonas [212] Coprococcus [212] Roseburia [212] |
Actinomyces [212] Dialister [212] Parabacteroides [212] |
Vitamin B2 | Alistipes [212] Blautia [212] Coprococcus [212] |
|
Vitamin B9 | Butyricicoccus [212] Roseburia [212] |
Alistipes [212] Anaerovorax [212] Megasphaera [212] Odoribacter [212] Parabacteroides [212] |
Vitamin C | Butyricicoccus [212] | Alistipes [212] Anaerovorax [212] Odoribacter [212] Parabacteroides [212] |
Vitamin D | Bacteroides [212,257] Clostridium [258] Lachnobacterum [259] Lactococcus [260] Prevotella [261] |
Bifidobacterium [257,261] Clostridium [257] Coprococcus [261] Dialister [212] Escherichia [262] Haemophilus [261] Lactococcus [259] Pseudomonas [262] Shigella [262] Veillonella [260,261] |
Vitamin E | Roseburia [212] | Acidaminococcus [212] Anaerotruncus [212] Odoribacter [212] |
Vitamin K | Butyricimonas [212] Coprococcus [212] |
Alistipes [212] Anaerotruncus [212] Fusobacteria [212] Parabacteroides [212] Ruminococcus [212] Streptococcus [212] |
Salt | Collinsella [212] Megamonas [212] Parabacteroides [212] |
Faecalibacterium [212] Lactobacillus [117] |
Polyphenols | Bacteroides [263,264] Bifidobacterum [263,265–267] Blautia [263] Clostridium [264] Eggerthella [263] Enterococcus [263] Eubacterium [263] Lactobacillus [264,266] Prevotella [263] Lactococcus [264] |
Blautia [268] Clostridium [266,268] |
Fibers | Akkermansia [268] Bacteroides [268] Clostridium [248] Coprococcus [248] Dorea [252] Haemophilus [248] Holdemania [248] Lactobacillus [268] Methanobrevibacter [252] Phascolarctobacterium [212] Roseburia [212,252] |
Acidaminococcus [212] Actinomyces [212] Anaerotruncus [212] Anaerovorax [212] Holdemania [212] Odoribacter [212] Parabacteroides [212] |
Short chain fatty acids | Barnesiella [212] Coprobacillus [212] Odoribacter [212] Parabacteroides [212] |
Prevotella [212] |
Long chain polyunsaturated fatty acids | Bacteroides [244] Bilophila [248] Coprococcus [244] Dorea [252] Holdemania [248] Lachnospira [244] Lactobacillus [252] Oscillospira [244] Roseburia [244,248] Ruminococcus [248] |
Butyricimonas [252] Collinsella [248] Slackia [252] Streptococcus [248] |
3.4. Strategies to study interplay between diet, gut microbiota, and MS
While diet and the gut microbiota have been investigated in isolation in the context of demyelinating disease, less well studied is the relationship between diet, gut microbiota, and disease. One approach to study this more complicated system is a top-down approach: to examine identified dietary risk factors for developing MS and test whether the effects of such a diet are mediated by the gut microbiota. This is the strategy employed by Wilck and colleagues [117] in which they examine a high salt diet and identify Lactobacillus murinis, a salt-sensitive gut microorganism that 1) was significantly depleted by a high salt diet, and 2) was found to suppress the Th17 response when used as a treatment in mice with EAE. Importantly, germ-free mice were utilized to isolate the effect of diet from the effect on the gut microbiota. Germ-free mice were fed a high salt diet, however this resulted in no change in Th17 frequency, compared to germ-free mice fed a normal salt diet, suggesting that gut microbiota are necessary for a high salt diet to induce Th17 cells [117]. Interestingly, this is seemingly at odds with previous studies indicating that salt can induce Th17 cells in vitro [115,116]. This strategy has also been used by Cignarella and colleagues [13] in which intermittent fasting was found to ameliorate EAE. They demonstrated that intermittent fasting changed the gut microbiota in mice and were able to recapitulate the benefit of intermittent fasting by colonizing germ-free mice with microbiota from intermittent fasting mice. Promisingly, in a pilot study in MS patients, they demonstrated that intermittent fasting also changed the gut microbiota of human patients comparable to mice at the phylum level [13].
Another approach to studying the relationship between diet, gut microbiota, and demyelinating disease is a bottom-up approach: starting with a microbial metabolite with known beneficial effects in EAE and determining whether supplementing the dietary precursor for the metabolite can ameliorate disease. This is the approach taken by Rothhammer and colleagues in which they identified gut microbiota-derived indole metabolites as a major beneficial microbial metabolite in EAE, previous work having identified tryptophan as the precursor for such indole metabolites [213], and then went on to demonstrate that mice with a tryptophan-deficient diet exhibited exacerbated EAE [214]. Critically, they show that antibiotic treatment reduced the level of indole-derived metabolites, suggesting that the gut microbiota contributes to the production of such metabolites [214]. However, it is unclear whether dietary tryptophan supplementation requires the gut microbiota to have a beneficial effect. In a prospective study of patients with pediatric-onset MS and CIS, higher levels of tryptophan and indole lactate were associated with a lower risk of MS [215]. Interestingly, a lower relative abundance of gut microbial genes involved in tryptophan catabolism was associated with increased relapse risk and a higher abundance of gut-microbe derived tryptophan metabolites was associated with lower disability [215]. It will be interesting to see whether supplementation with tryptophan or indole would be beneficial for patients with MS.
Another approach to studying diet, gut microbiota, and demyelinating disease is an unbiased approach: screening diets, using the EAE model, that modulate disease regardless of prior association with MS, sequencing the gut microbiota of mice on different diets, and subsequently validating the functional impact of the gut microbiota. This is the approach we took when we observed that mice on different diets exhibited different disease [216]. While the two diets were considered standard rodent diets, they differed in macro-and micro-nutrient composition (specific nutritional information can be found in [216]) with the beneficial diet supplemented with vitamins to accommodate autoclave sterilization and the detrimental diet formulated to exclude soybean protein. Mice on the beneficial diet had lower clinical scores, lower incidence of disease, less weight loss, and lower mortality compared to mice on the alternative diet. Spinal cord pathology also showed that mice on the detrimental diet had higher pathology scores for demyelination, perivascular cuffing, meningitis, and overall pathology. Importantly, the severity of disease did not cluster in individual cages of a particular diet, making the possibility of cage-specific effects less likely. To determine the microbiota changes due to diet and disease, we performed 16S rRNA sequencing on mice grouped by clinical score and diet. We also analyzed the microbial composition of naive mice and mice administered complete Freund’s adjuvant and Bordetella pertussis toxin without myelin peptide to control for changes in microbiota due to the injection adjuvant. Sequencing the gut microbiota of mice on these different diets and accounting for the severity of disease exhibited by individual mice, we identified Lactobacillus species as being depleted in mice induced with EAE compared to naive and adjuvant injected control animals, inversely correlated in terms of relative abundance and EAE severity, and enriched in EAE-induced animals on the beneficial diet compared to EAE-induced animals on the detrimental diet [181]. The Lactobacillus genus was unique in exhibiting these three features. Finally, we were able to recapitulate the effects of the beneficial diet by supplementing a human gut-derived commensal strain of Lactobacillus [181]. In our experimental paradigm, mice on the beneficial diet were fed L. paracasei daily via oral gavage starting two weeks prior to EAE induction. L. paracasei-treated mice exhibited lower clinical scores, lower EAE incidence, and less weight loss compared to vehicle-treated animals. While this approach has the ability to uncover novel relationships between diet, gut microbiota, and disease, the complex relationship between these factors makes untangling the mechanisms of the system difficult.
4. Discussion
Whether using the top-down, bottom-up, or unbiased approach to dissect diet, microbiota, and disease relationships, it is clear that such networks are complex. The difficulty in understanding the interplay between these factors is magnified by tentative results of diet and probiotic treatments in MS patients that require validation in larger, longer clinical trials. Outstanding questions in this area are: Does the benefit of vitamin D supplementation, the most studied dietary intervention in MS, work through a microbiota-based mechanism? How stable are diet-induced changes in gut microbiota? How are diet-based and gut microbiota-based interventions affected by current MS disease modifying therapies? Addressing these questions will require the use of 16S rRNA sequencing and bioinformatics tools to assess changes in the gut microbiota, germ-free animal systems to isolate the effects of diet versus the gut microbiota, and the commitment of clinician scientists to pursue well-powered and longitudinal clinical trials of these interventions.
Acknowledgements
This work was supported by the National Institutes of Health 5R01NS065714, 1R01NS091939 and 5T32AI055434 (J.M.S.S.).
References
- [1].Reich DS, Lucchinetti CF, Calabresi PA, Multiple sclerosis. Longo DL, editor, N. Engl. J. Med 378 (2018) 169–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Farinotti M, Vacchi L, Simi S, Di Pietrantonj C, Brait L, Filippini G, Dietary interventions for multiple sclerosis. Cochrane Multiple Sclerosis and Rare Diseases of the CNS Group, Cochrane Database Syst. Rev 12 (2012) John Wiley & Sons, Ltd. (CD004192). [DOI] [PubMed] [Google Scholar]
- [3].Jagannath VA, Fedorowicz Z, Asokan GV, Robak EW, Whamond L, Vitamin D for the management of multiple sclerosis. Cochrane Multiple Sclerosis and Rare Diseases of the CNS Group, Cochrane Database Syst. Rev 74 (2010) John Wiley & Sons, Ltd. (CD008422). [DOI] [PubMed] [Google Scholar]
- [4].Riccio P, Rossano R, Nutrition facts in multiple sclerosis, ASN Neuro, vol. 7, SAGE Publications, Los Angeles, CA, 2015(175909141456818). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Swank RL, Multiple sclerosis; a correlation of its incidence with dietary fat, Am. J. Med. Sci 220 (1950) 421–430. [PubMed] [Google Scholar]
- [6].Swank RL, Dugan BB, Effect of low saturated fat diet in early and late cases of multiple sclerosis, Lancet 336 (1990) 37–39. [DOI] [PubMed] [Google Scholar]
- [7].Weindruch R, Walford RL, Fligiel S, Guthrie D, The retardation of aging in mice by dietary restriction: longevity, cancer, immunity and lifetime energy intake, J. Nutr 116 (1986) 641–654. [DOI] [PubMed] [Google Scholar]
- [8].Zainal TA, Oberley TD, Allison DB, Szweda LI, Weindruch R, Caloric restriction of rhesus monkeys lowers oxidative damage in skeletal muscle, FASEB J 14 (2000) 1825–1836 Federation of American Societies for Experimental Biology. [DOI] [PubMed] [Google Scholar]
- [9].Higami Y, Barger JL, Page GP, Allison DB, Smith SR, Prolla TA, et al. , Energy restriction lowers the expression of genes linked to inflammation, the cytoskeleton, the extracellular matrix, and angiogenesis in mouse adipose tissue, J. Nutr 136 (2006) 343–352. [DOI] [PubMed] [Google Scholar]
- [10].Piccio L, Stark JL, Cross AH, Chronic calorie restriction attenuates experimental autoimmune encephalomyelitis, J. Leukoc. Biol 84 (2008) 940–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Kafami L, Raza M, Razavi A, Mirshafiey A, Movahedian M, Khorramizadeh MR, Intermittent feeding attenuates clinical course of experimental autoimmune encephalomyelitis in C57BL/6 mice, Avicenna J. Med. Biotechnol 2 (2010) 47–52. Avicenna Research Institute. [PMC free article] [PubMed] [Google Scholar]
- [12].Esquifino AI, Cano P, Jimenez-Ortega V, Fernández-Mateos MP, Cardinali DP, Immune response after experimental allergic encephalomyelitis in rats subjected to calorie restriction, J. Neuroinflammation 4 (2007) 6 BioMed Central. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Cignarella F, Cantoni C, Ghezzi L, Salter A, Dorsett Y, Chen L, et al. , Intermittent fasting confers protection in CNS autoimmunity by altering the gut microbiota, Cell Metab 27 (2018) 1222–1226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Choi IY, Piccio L, Childress P, Bollman B, Ghosh A, Brandhorst S, et al A diet mimicking fasting promotes regeneration and reduces autoimmunity and multiple sclerosis symptoms, Cell Reports 15 (2016) 2136–2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Kossoff EH, McGrogan JR, Worldwide use of the ketogenic diet, Epilepsia, 46 John Wiley & Sons, Ltd; (10.1111), 2005, pp. 280–289. [DOI] [PubMed] [Google Scholar]
- [16].Kim DY, Hao J, Liu R, Turner G, Shi F-D, Rho JM, Inflammation-mediated memory dysfunction and effects of a ketogenic diet in a murine model of multiple sclerosis. Villoslada P, editor, PLoS One 7 (2012) e35476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Brenton JN, Banwell B, Bergqvist AGC, Lehner-Gulotta D, Gampper L, Leytham E, et al. , Pilot study of a ketogenic diet in relapsing-remitting MS, Neurol. Neuroimmunol. Neuroinflamm 6 (2019) e565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Hruby A, Hu FB, The epidemiology of obesity: a big picture, Pharmacoeconomics 33 (2015) 673–689 Springer International Publishing. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].GBD 2016 Multiple Sclerosis Collaborators, Global, regional, and national burden of multiple sclerosis 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016, Lancet Neurol 18 (2019) 269–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Statovci D, Aguilera M, MacSharry J, Melgar S, The impact of Western diet and nutrients on the microbiota and immune response at mucosal interfaces, Front. Immunol 8 (2017) 838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Timmermans S, Bogie JFJ, Vanmierlo T, Lütjohann D, Stinissen P, Hellings N, et al. , High fat diet exacerbates neuroinflammation in an animal model of multiple sclerosis by activation of the renin angiotensin system, J. Neuroimmune Pharmacol 9 (2014) 209–217 Springer LJS. [DOI] [PubMed] [Google Scholar]
- [22].Haghikia A, Jörg S, Duscha A, Berg J, Manzel A, Waschbisch A, et al. , Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine, Immunity 43 (2015) 817–829. [DOI] [PubMed] [Google Scholar]
- [23].Winer S, Paltser G, Chan Y, Tsui H, Engleman E, Winer D, et al. , Obesity predisposes to Th17 bias, Eur. J. Immunol 39 (2009) 2629–2635 John Wiley & Sons, Ltd. [DOI] [PubMed] [Google Scholar]
- [24].Hasan M, Seo J-E, Rahaman KA, Min H, Kim KH, Park J-H, et al. , Novel genes in brain tissues of EAE-induced normal and obese mice: upregulation of metal ion-binding protein genes in obese-EAE mice, Neuroscience 343 (2017) 322–336. [DOI] [PubMed] [Google Scholar]
- [25].Hedström AK, Olsson T, Alfredsson L, High body mass index before age 20 is associated with increased risk for multiple sclerosis in both men and women, Mult. Scler 18 (2012) 1334–1336. [DOI] [PubMed] [Google Scholar]
- [26].Munger KL, Chitnis T, Ascherio A, Body size and risk of MS in two cohorts of US women, Neurology 73 (2009) 1543–1550 Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Wesnes K, Riise T, Casetta I, Drulovic J, Granieri E, Holmpy T, et al. , Body size and the risk of multiple sclerosis in Norway and Italy: the EnvIMS study, Mult. Scler 21 (2015) 388–395. [DOI] [PubMed] [Google Scholar]
- [28].Langer-Gould A, Brara SM, Beaber BE, Koebnick C, Childhood obesity and risk of pediatric multiple sclerosis and clinically isolated syndrome, Neurology 80 (2013) 548–552 Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Watzl B, Kulling SE, Möseneder J, Barth SW, Bub A, A 4-wk intervention with high intake of carotenoid-rich vegetables and fruit reduces plasma C-reactive protein in healthy, nonsmoking men, Am. J. Clin. Nutr 82 (2005) 1052–1058. [DOI] [PubMed] [Google Scholar]
- [30].Yadav V, Marracci G, Kim E, Spain R, Cameron M, Overs S, et al. , Low-fat, plant-based diet in multiple sclerosis: a randomized controlled trial, Mult. Scler. Relat. Disord 9 (2016) 80–90. [DOI] [PubMed] [Google Scholar]
- [31].Zhang C, Björkman A, Cai K, Liu G, Wang C, Li Y, et al. , Impact of a 3-months vegetarian diet on the gut microbiota and immune repertoire, Front. Immunol 9 (2018) 908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Trichopoulou A, Costacou T, Bamia C, Trichopoulos D, Adherence to a Mediterranean diet and survival in a Greek population, N. Engl. J. Med 348 (2003) 2599–2608 Massachusetts Medical Society. [DOI] [PubMed] [Google Scholar]
- [33].Casas R, Sacanella E, Estruch R, The immune protective effect of the Mediterranean diet against chronic low-grade inflammatory diseases, Endocr. Metab. Immune Disord. Drug Targets 14 (2014) 245–254 Bentham Science Publishers. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Ambring A, Johansson M, Axelsen M, Gan L, Strandvik B, Friberg P, Mediterranean-inspired diet lowers the ratio of serum phospholipid n-6 to n-3 fatty acids, the number of leukocytes and platelets, and vascular endothelial growth factor in healthy subjects, Am. J. Clin. Nutr 83 (2006) 575–581. [DOI] [PubMed] [Google Scholar]
- [35].Black LJ, Baker K, Ponsonby A-L, van der Mei I, Lucas RM, Pereira G, et al. , 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 149 (2019) 1385–1392. [DOI] [PubMed] [Google Scholar]
- [36].Sedaghat F, Jessri M, Behrooz M, Mirghotbi M, Rashidkhani B, Mediterranean diet adherence and risk of multiple sclerosis: a case-control study, Asia Pac. J. Clin. Nutr 25 (2016) 377–384 HEC Press. [DOI] [PubMed] [Google Scholar]
- [37].Rotstein DL, Cortese M, Fung TT, Chitnis T, Ascherio A, Munger KL, Diet quality and risk of multiple sclerosis in two cohorts of US women, Mult. Scler 15 (2018) (1352458518807061). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Sand IK, Benn EK, Fabian M, Fitzgerald KC, Digga E, Deshpande R, et al. , Randomized-controlled trial of a modified Mediterranean dietary program for multiple sclerosis: a pilot study, Mult. Scler. Relat. Disord 36 (2019) 101403 Elsevier. (101403). [DOI] [PubMed] [Google Scholar]
- [39].Correale J, Ysrraelit MC, Gaitan MI, Immunomodulatory effects of vitamin D in multiple sclerosis, Brain 132 (2009) 1146–1160. [DOI] [PubMed] [Google Scholar]
- [40].Swank RL, Lerstad O, Strom A, Backer J, Multiple sclerosis in rural Norway its geographic and occupational incidence in relation to nutrition, N. Engl. J. Med 246 (1952) 722–728. [DOI] [PubMed] [Google Scholar]
- [41].van der Mei IA, Ponsonby AL, Blizzard L, Dwyer T, Regional variation in multiple sclerosis prevalence in Australia and its association with ambient ultra-violet radiation, Neuroepidemiology 20 (2001) 168–174 Karger Publishers. [DOI] [PubMed] [Google Scholar]
- [42].Vukusic S, Van Bockstael V, Gosselin S, Confavreux C, Regional variations in the prevalence of multiple sclerosis in French farmers, J. Neurol. Neurosurg. Psychiatry 78 (2007) 707–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].International Multiple Sclerosis Genetics Consortium, Wellcome Trust Case Control Consortium 2, Sawcer S, Hellenthal G, Pirinen M, CCA Spencer, et al. , Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis, Nature 476 (2011) 214–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Lemire JM, Archer DC, 1,25-dihydroxyvitamin D3 prevents the in vivo induction of murine experimental autoimmune encephalomyelitis, J. Clin. Invest 87 (1991) 1103–1107. American Society for Clinical Investigation. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Cantorna MT, Hayes CE, DeLuca HF, 1,25-Dihydroxyvitamin D3 reversibly blocks the progression of relapsing encephalomyelitis, a model of multiple sclerosis, Proc. Natl. Acad. Sci 93 (1996) 7861–7864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Chiuso-Minicucci F, Ishikawa LLW, Mimura LAN, de Fraga-Silva TFC, Franca TGD, Zorzella-Pezavento SFG, et al. , Treatment with vitamin D/MOG association suppresses experimental autoimmune encephalomyelitis. Forsthuber T, editor, PLoS One 10 (2015) (eOl 25836–14). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Lemire JM, Adams JS, Sakai R, Jordan SC, 1 alpha,25-dihydroxyvitamin D3 suppresses proliferation and immunoglobulin production by normal human peripheral blood mononuclear cells, J. Clin. Invest 74 (1984) 657–661. American Society for Clinical Investigation. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Chen S, Sims GP, Chen XX, Gu YY, Chen S, Lipsky PE, Modulatory effects of 1,25-dihydroxyvitamin D3 on human B cell differentiation, J. Immunol 179 (2007) 1634–1647. [DOI] [PubMed] [Google Scholar]
- [49].Bhalla AK, Amen to EP, Serog B, Glimcher LH, 1,25-Dihydroxyvitamin D3 inhibits antigen-induced T cell activation, J. Immunol 133 (1984) 1748–1754. [PubMed] [Google Scholar]
- [50].Mattner F, Smiroldo S, Galbiati F, Muller M, Di Lucia P, Poliani PL, et al. , Inhibition of Th1 development and treatment of chronic-relapsing experimental allergic encephalomyelitis by a non-hypercalcemic analogue of 1,25-dihydrox-yvitamin D(3), Eur. J. Immunol 30 (2000) 498–508. [DOI] [PubMed] [Google Scholar]
- [51].Tang J, Zhou R, Luger D, Zhu W, Silver PB, Grajewski RS, et al. , Calcitriol suppresses antiretinal autoimmunity through inhibitory effects on the Th17 effector response, J. Immunol 182 (2009) 4624–4632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Penna G, Roncari A, Amuchastegui S, Daniel KC, Berti E, Colonna M, et al. , Expression of the inhibitory receptor ILT3 on dendritic cells is dispensable for induction of CD4 + Foxp3 + regulatory T cells by 1,25-dihydroxyvitamin D3, Blood 106 (2005) 3490–3497. American Society of Hematology. [DOI] [PubMed] [Google Scholar]
- [53].Piemonti L, Monti P, Sironi M, Fraticelli P, Leone BE, Dal Cin E, et al. , Vitamin D3 affects differentiation, maturation, and function of human monocyte-derived dendritic cells, J. Immunol 164 (2000) 4443–4451. [DOI] [PubMed] [Google Scholar]
- [54].Griffin MD, Lutz W, Phan VA, Bachman LA, McKean DJ, Kumar R, Dendritic cell modulation by 1 alpha,25 dihydroxyvitamin D3 and its analogs: a vitamin D receptor-dependent pathway that promotes a persistent state of immaturity in vitro and in vivo, Proc. Natl. Acad. Sci 98 (2001) 6800–6805 National Academy of Sciences. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Széles L, Keresztes G, Töröcsik D, Balajthy Z, Krenács L, Póliska S, et al. , 1,25-dihydroxyvitamin D3 is an autonomous regulator of the transcriptional changes leading to a tolerogenic dendritic cell phenotype, J. Immunol 182 (2009) 2074–2083. [DOI] [PubMed] [Google Scholar]
- [56].Wang Y, Marling SJ, Zhu JG, Severson KS, DeLuca HF, Development of experimental autoimmune encephalomyelitis (EAE) in mice requires vitamin D and the vitamin D receptor, Proc. Natl. Acad. Sci. U. S. A 109 (2012) 8501–8504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].von Essen MR, Kongsbak M, Schjerling P, Olgaard K, Odum N, Geisler C, Vitamin D controls T cell antigen receptor signaling and activation of human T cells, Nat. Publ. Group 11 (2010) 344–349. [DOI] [PubMed] [Google Scholar]
- [58].Mirzaei F, Michels KB, Munger K, O’Reilly E, Chitnis T, Forman MR, et al. , Gestational vitamin D and the risk of multiple sclerosis in offspring, Ann. Neurol 70 (2011) 30–40 John Wiley & Sons, Ltd. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Ascherio A, Munger KL, White R, Köchert K, Simon KC, Polman CH, et al. , Vitamin D as an early predictor of multiple sclerosis activity and progression, JAMA Neurol 71 (2014) 306–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Fitzgerald KC, Munger KL, Kochert K, Arnason BGW, Co mi G, Cook S, et al. , Association of vitamin D Levels with multiple sclerosis activity and progression in patients receiving interferon Beta-lb, JAMA Neurol 72 (2015) 1458–1465. American Medical Association. [DOI] [PubMed] [Google Scholar]
- [61].Sotirchos ES, Bhargava P, Eckstein C, Van Haren K, Baynes M, Ntranos A, et al. , Safety and immunologic effects of high-vs low-dose cholecalciferol in multiple sclerosis, Neurology 86 (2016) 382–390 Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Burton JM, Kimball S, Vieth R, Bar-Or A, Dosch H-M, Cheung R, et al A phase I/II dose-escalation trial of vitamin D3 and calcium in multiple sclerosis, Neurology 74 (2010) 1852–1859 Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Ueda P, Rafatnia F, Bäärnhielm M, Fröbom R, Korzunowicz G, Lönnerbro R, et al. , Neonatal vitamin D status and risk of multiple sclerosis, Ann. Neurol 76 (2014) 338–346 John Wiley & Sons, Ltd. [DOI] [PubMed] [Google Scholar]
- [64].Stein MS, Liu Y, Gray OM, Baker JE, Kolbe SC, Ditchfield MR, et al A randomized trial of high-dose vitamin D2 in relapsing-remitting multiple sclerosis, Neurology 77 (2011) 1611–1618 Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology. [DOI] [PubMed] [Google Scholar]
- [65].James E, Dobson R, Kuhle J, Baker D, Giovannoni G, Ramagopalan SV, The effect of vitamin D-related interventions on multiple sclerosis relapses: a meta-analysis, Mult Scler, 3rd ed., vol. 19, SAGE Publications, London, England, 2013, pp. 1571–1579. [DOI] [PubMed] [Google Scholar]
- [66].Wiedermann U, Hanson LA, Kahu H, Dahlgren UI, Aberrant T-cell function in vitro and impaired T-cell dependent antibody response in vivo in vitamin A-deficient rats, Immunology 80 (1993) 581–586 Wiley-Blackwell. [PMC free article] [PubMed] [Google Scholar]
- [67].Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M, et al. , Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid, Science 317 (2007) 256–260. [DOI] [PubMed] [Google Scholar]
- [68].Benson MJ, Pino-Lagos K, Rosemblatt M, Noelle RJ, All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation, J. Exp. Med 204 (2007) 1765–1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Uematsu S, Fujimoto K, Jang MH, Yang B-G, Jung Y-J, Nishiyama M, et al. , Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing toll-like receptor 5, Nat. Publ. Group 9 (2008) 769–776. [DOI] [PubMed] [Google Scholar]
- [70].Massacesi L, Castigli E, Vergelli M, Olivotto J, Abbamondi AL, Sarlo F, et al. , Immunosuppressive activity of 13-cis-retinoic acid and prevention of experimental autoimmune encephalomyelitis in rats, J. Clin. Invest 88 (1991) 1331–1337. American Society for Clinical Investigation. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Racke MK, Burnett D, Pak SH, Albert PS, Cannella B, Raine CS, et al. , Retinoid treatment of experimental allergic encephalomyelitis. IL-4 production correlates with improved disease course, J. Immunol 154 (1995) 450–458. [PubMed] [Google Scholar]
- [72].Zhan X-X, Liu Y, Yang J-F, Wang G-Y, Mu L, Zhang T-S, et al. , All-trans-retinoic acid ameliorates experimental allergic encephalomyelitis by affecting dendritic cell and monocyte development, Immunology, 5th ed., vol. 138, John Wiley & Sons, Ltd; (10.1111), 2013, pp. 333–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Mukherjee S, Date A, Patravale V, Korting HC, Roeder A, Weindl G, Retinoids in the treatment of skin aging: an overview of clinical efficacy and safety, Clin. Interv. Aging 1 (2006) 327–348 Dove Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Besler HT, Comoğlu S, okçu Z, Serum levels of antioxidant vitamins and lipid peroxidation in multiple sclerosis, Nutr. Neurosci 5 (2002) 215–220. [DOI] [PubMed] [Google Scholar]
- [75].Royal W, Gartner S, Gajewski CD, Retinol measurements and retinoid receptor gene expression in patients with multiple sclerosis, Mult. Scler 8 (2002) 452–458. [DOI] [PubMed] [Google Scholar]
- [76].Løken-Amsrud KI, Myhr K-M, Bakke SJ, Beiske AG, Bjerve KS, Bjørnarå BT, et al. , Retinol levels are associated with magnetic resonance imaging outcomes in multiple sclerosis, Mult Scler, vol. 19, SAGE Publications, London, England, 2013, pp. 451–457. [DOI] [PubMed] [Google Scholar]
- [77].Qu ZX, Dayal A, Jensen MA, Arnason BG, All-trans retinoic acid potentiates the ability of interferon beta-lb to augment suppressor cell function in multiple sclerosis, Arch. Neurol 55 (1998) 315–321. [DOI] [PubMed] [Google Scholar]
- [78].Runia TF, Hop WCJ, de Rijke YB, Hintzen RQ, Vitamin a is not associated with exacerbations in multiple sclerosis, Mult. Scler. Relat. Disord 3 (2014) 34–39. [DOI] [PubMed] [Google Scholar]
- [79].Evans E, Piccio L, Cross AH, Use of vitamins and dietary supplements by patients with multiple sclerosis: a review, JAMA Neurol 75 (2018) 1013–1021. [DOI] [PubMed] [Google Scholar]
- [80].Khosravi-Largani M, Pourvali-Talatappeh P, Rousta AM, Karimi-Kivi M, Noroozi E, Mahjoob A, et al. , A review on potential roles of vitamins in incidence, progression, and improvement of multiple sclerosis, eNeurologicalSci 10 (2018) 37–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Costantini A, Nappo A, Pala MI, Zappone A, High dose thiamine improves fatigue in multiple sclerosis, BMJ Case Rep 2013 (2013) (bcr2013009144–4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Ogunleye AJ, Odutuga AA, The effect of riboflavin deficiency on cerebrum and cerebellum of developing rat brain, J. Nutr. Sci. Vitaminol 35 (1989) 193–197 Center for Academic Publications Japan. [DOI] [PubMed] [Google Scholar]
- [83].Naghashpour M, Amani R, Sarkaki A, Ghadiri A, Samarbafzadeh A, Jafarirad S, et al. , Brain-derived neurotrophic and immunologic factors: beneficial effects of riboflavin on motor disability in murine model of multiple sclerosis, Iran J. Basic Med. Sci 19 (2016) 439–448 Mashhad University of Medical Sciences. [PMC free article] [PubMed] [Google Scholar]
- [84].Ghadirian P, Jain M, Ducic S, Shatenstein B, Morisset R, Nutritional factors in the aetiology of multiple sclerosis: a case-control study in Montreal, Canada, Int. J. Epidemiol 27 (1998) 845–852. [DOI] [PubMed] [Google Scholar]
- [85].Naghashpour M, Majdinasab N, Shakerinejad G, Kouchak M, Haghighizadeh MH, Jarvandi F, et al. , Riboflavin supplementation to patients with multiple sclerosis does not improve disability status nor is riboflavin supplementation correlated to homocysteine, Int. J. Vitam. Nutr. Res 83 (2013) 281–290 Verlag Hans Huber. [DOI] [PubMed] [Google Scholar]
- [86].Braidy N, Berg J, Clement J, Khorshidi F, Poljak A, Jayasena T, et al. , Role of nicotinamide adenine dinucleotide and related precursors as therapeutic targets for age-related degenerative diseases: rationale, biochemistry, pharmacokinetics, and outcomes, Antioxid. Redox Signal 30 (2019) 251–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Zhang J, Chen J, Li Y, Cui X, Zheng X, Roberts C, et al. , Niaspan treatment improves neurological functional recovery in experimental autoimmune encephalomyelitis mice, Neurobiol. Dis 32 (2008) 273–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Parra M, Stahl S, Hellmann H, Vitamin B6 and its role in cell metabolism and physiology, Cells 7 (2018) 84 Multidisciplinary Digital Publishing Institute. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Wetzels S, Wouters K, Miyata T, Scheijen JLJM, Hendriks JJA, Schalkwijk CG, et al. , Advanced glycation endproducts are increased in the animal model of multiple sclerosis but cannot be reduced by pyridoxamine treatment or glyoxalase 1 overexpression, Int. J. Mol. Sci 19 (2018) 1311 Multidisciplinary Digital Publishing Institute. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Sedel F, Bernard D, Mock DM, Tourbah A, Targeting demyelination and virtual hypoxia with high-dose biotin as a treatment for progressive multiple sclerosis, Neuropharmacology 110 (2016) 644–653. [DOI] [PubMed] [Google Scholar]
- [91].Buonvicino D, Ranieri G, Pratesi S, Guasti D, Chiarugi A, Neuroimmunological characterization of a mouse model of primary progressive experimental autoimmune encephalomyelitis and effects of immunosuppressive or neuroprotective strategies on disease evolution, Exp. Neurol 322 (2019) 113065. [DOI] [PubMed] [Google Scholar]
- [92].Sedel F, Papeix C, Bellanger A, Touitou V, Lebrun-Frenay C, Galanaud D, et al. , High doses of biotin in chronic progressive multiple sclerosis: a pilot study, Mult. Scler. Relat. Disord 4 (2015) 159–169. [DOI] [PubMed] [Google Scholar]
- [93].Tourbah A, Lebrun-Frenay C, Edan G, Clanet M, Papeix C, Vukusic S, et al. , MD1003 (high-dose biotin) for the treatment of progressive multiple sclerosis: a randomised, double-blind, placebo-controlled study, Mult Scler, vol. 22, SAGE Publications, London, England, 2016, pp. 1719–1731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Shane B, Folate and vitamin B12 metabolism: overview and interaction with riboflavin, vitamin B6, and polymorphisms, Food Nutr Bull, 3rd ed., 29 SAGE Publications, Los Angeles, CA, 2008(S5–16–discussionS17–9). [DOI] [PubMed] [Google Scholar]
- [95].Mastronardi FG, Min W, Wang H, Winer S, Dosch M, Boggs JM, et al. , Attenuation of experimental autoimmune encephalomyelitis and nonimmune demyelination by IFN-beta plus vitamin B12: treatment to modify notch-l/sonic hedgehog balance, J. Immunol 172 (2004) 6418–6426. American Association of Immunologists. [DOI] [PubMed] [Google Scholar]
- [96].Zhu Y, He Z-Y, Liu H-N, Meta-analysis of the relationship between homocysteine, vitamin B12, folate, and multiple sclerosis, J. Clin. Neurosci 18 (2011) 933–938. [DOI] [PubMed] [Google Scholar]
- [97].Scalabrino G, Veber D, De Giuseppe R, Roncaroli F, Low levels of cobalamin, epidermal growth factor, and normal prions in multiple sclerosis spinal cord, Neuroscience 298 (2015) 293–301. [DOI] [PubMed] [Google Scholar]
- [98].Nozari E, Ghavamzadeh S, Razazian N, The effect of vitamin B12 and folic acid supplementation on serum homocysteine, anemia status and quality of life of patients with multiple sclerosis, Clin. Nutr. Res 8 (2019) 36–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Yoshii K, Hosomi K, Sawane K, Kunisawa J, Metabolism of dietary and microbial vitamin B family in the regulation of host immunity, Front. Nutr 6 (2019) 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Naidu KA, Vitamin C in human health and disease is still a mystery? An overview, Nutr J. BioMed. Central 2 (2003) 7–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Spitsin SV, Scott GS, Mikheeva T, Zborek A, Kean RB, Brimer CM, et al. , Comparison of uric acid and ascorbic acid in protection against EAE, Free Radic. Biol. Med 33 (2002) 1363–1371. [DOI] [PubMed] [Google Scholar]
- [102].Guo Y-E, Suo N, Cui X, Yuan Q, Xie X, Vitamin C promotes oligodendrocytes generation and remyelination, Glia 66 (2018) 1302–1316 John Wiley & Sons, Ltd. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Sen CK, Khanna S, Roy S, Tocotrienols: vitamin E beyond tocopherols, Life Sci 78 (2006) 2088–2098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Xue H, Ren H, Zhang L, Sun X, Wang W, Zhang S, et al. , Alpha-tocopherol ameliorates experimental autoimmune encephalomyelitis through the regulation of Th1 cells, Iran J. Basic Med. Sci 19 (2016) 561–566 Mashhad University of Medical Sciences. [PMC free article] [PubMed] [Google Scholar]
- [105].Blanchard B, Heurtaux T, Garcia C, Moll NM, Caillava C, Grandbarbe L, et al. , Tocopherol derivative TFA-12 promotes myelin repair in experimental models of multiple sclerosis, J. Neurosci. Soc. Neurosci 33 (2013) 11633–11642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Goudarzvand M, Javan M, Mirnajafi-Zadeh J, Mozafari S, Tiraihi T, Vitamins E and D3 attenuate demyelination and potentiate remyelination processes of hip-pocampal formation of rats following local injection of ethidium bromide, Cell. Mol. Neurobiol 30 (2010) 289–299 6 ed.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Løken-Amsrud KI, Myhr K-M, Bakke SJ, Beiske AG, Bjerve KS, Bjørnarå BT, et al. , Alpha-tocopherol and MRI outcomes in multiple sclerosis-association and prediction. Oreja-Guevara C, editor, PLoS One 8 (2013) Public Library of Science. (e54417). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Card DJ, Gorska R, Cutler J, Harrington DJ, Vitamin K metabolism: current knowledge and future research. Traber MG, editor, Mol. Nutr. Food Res 58 (2014) 1590–1600 John Wiley & Sons, Ltd. [DOI] [PubMed] [Google Scholar]
- [109].Moriya M, Nakatsuji Y, Okuno T, Hamasaki T, Sawada M, Sakoda S, Vitamin K2 ameliorates experimental autoimmune encephalomyelitis in Lewis rats, J. Neuroimmunol 170 (2005) 11–20. [DOI] [PubMed] [Google Scholar]
- [110].Popescu DC, Huang H, Singhal NK, Shriver L, McDonough J, Clements RJ, et al. , Vitamin K enhances the production of brain sulfatides during remyelination. de Castro F, editor, PLoS One 13 (2018) eO203057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Lasemi R, Kundi M, Moghadam NB, Moshammer H, Hainfellner JA, Vitamin K2 in multiple sclerosis patients, Wien. Klin. Wochenschr 130 (2018) 307–313 4 ed. Springer Vienna. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Brown IJ, Tzoulaki I, Candeias V, Elliott P, Salt intakes around the world: implications for public health, Int. J. Epidemiol 38 (2009) 791–813. [DOI] [PubMed] [Google Scholar]
- [113].Junger WG, Liu FC, Loomis WH, Hoyt DB, Hypertonic saline enhances cellular immune function, Circ. Shock 42 (1994) 190–196. [PubMed] [Google Scholar]
- [114].Shapiro L, Dinarello CA, Osmotic regulation of cytokine synthesis in vitro, Proc. Natl. Acad. Sci 92 (1995) 12230–12234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Wu C, Yosef N, Thalhamer T, Zhu C, Xiao S, Kishi Y, et al. , Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1, Nature 496 (2013) 513–517 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Kleinewietfeld M, Manzel A, Titze J, Kvakan H, Yosef N, Linker RA, et al. , Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells, Nature 496 (2013) 518–522 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus H, et al. , Salt-responsive gut commensal modulates TH17 axis and disease, Nature 551 (2017) 585–589 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Farez MF, Fiol MP, Gaitán MI, Quintana FJ, Correale J, Sodium intake is associated with increased disease activity in multiple sclerosis, J. Neurol. Neurosurg. Psychiatry 86 (2015) 26–31. [DOI] [PubMed] [Google Scholar]
- [119].Fitzgerald KC, Munger KL, Hartung H-P, Freedman MS, Montalban X, Edan G, et al. , Sodium intake and multiple sclerosis activity and progression in BENEFIT, Ann. Neurol 82 (2017) 20–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Cortese M, Yuan C, Chitnis T, A, Ascherio, K.L. Munger, No association between dietary sodium intake and the risk of multiple sclerosis, Neurology 89 (2017) 1322–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].McDonald J, Graves J, Waldman A, Lotze T, Schreiner T, Belman A, et al A case-control study of dietary salt intake in pediatric-onset multiple sclerosis, Mult. Scler. Relat. Disord 6 (2016) 87–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Nourbakhsh B, Graves J, Casper TC, Lulu S, Waldman A, Belman A, et al. , Dietary salt intake and time to relapse in paediatric multiple sclerosis, J. Neurol. Neurosurg. Psychiatry 87 (2016) 1350–1353 BMJ Publishing Guoup Ltd. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Pandey KB, Rizvi SI, Plant polyphenols as dietary antioxidants in human health and disease, Oxid. Med. Cell. Longev 2 (2009) 270–278 Hindawi. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Ruiz PA, Haller D, Functional diversity of flavonoids in the inhibition of the proinflammatory NF-kappaB, IRF, and Akt signaling pathways in murine intestinal epithelial cells, J. Nutr 136 (2006) 664–671. [DOI] [PubMed] [Google Scholar]
- [125].Aktas O, Prozorovski T, Smorodchenko A, Savaskan NE, Lauster R, Kloetzel P-M, et al. , Green tea epigallocatechin-3-gallate mediates T cellular NF-kappa B inhibition and exerts neuroprotection in autoimmune encephalomyelitis, J. Immunol 173 (2004) 5794–5800. American Association of Immunologists. [DOI] [PubMed] [Google Scholar]
- [126].Ginwala R, McTish E, Raman C, Singh N, Nagarkatti M, Nagarkatti P, et al. , Apigenin, a natural flavonoid, attenuates EAE severity through the modulation of dendritic cell and other immune cell functions, J. Neuroimmune Pharmacol 11 (2016) 36–47 Springer US. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Haghmorad D, Mahmoudi MB, Salehipour Z, Jalayer Z, Momtazi Brojeni AA, Rastin M, et al. , Hesperidin ameliorates immunological outcome and reduces neuroinflammation in the mouse model of multiple sclerosis, J. Neuroimmunol 302 (2017) 23–33. [DOI] [PubMed] [Google Scholar]
- [128].Hendriks JJA, Alblas J, van der Pol SMA, van Tol EAF, Dijkstra CD, de Vries HE, Flavonoids influence monocytic GTPase activity and are protective in experimental allergic encephalitis, J. Exp. Med 200 (2004) 1667–1672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Ghaiad HR, Nooh MM, El-Sawalhi MM, Shaheen AA, Resveratrol promotes remyelination in cuprizone model of multiple sclerosis: biochemical and histological study, Mol. Neurobiol 54 (2017) 3219–3229. [DOI] [PubMed] [Google Scholar]
- [130].Sato F, Martinez NE, Shahid M, Rose JW, Carlson NG, Tsunoda I, Resveratrol exacerbates both autoimmune and viral models of multiple sclerosis, Am. J. Pathol 183 (2013) 1390–1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Verbeek R, van Tol EAF, van Noort JM, Oral flavonoids delay recovery from experimental autoimmune encephalomyelitis in SJL mice, Biochem. Pharmacol 70 (2005) 220–228. [DOI] [PubMed] [Google Scholar]
- [132].Morini M, Roccatagliata L, Deh’Eva R, Pedemonte E, Furlan R, Minghelli S, et al. , Alpha-lipoic acid is effective in prevention and treatment of experimental autoimmune encephalomyelitis, J. Neuroimmunol 148 (2004) 146–153. [DOI] [PubMed] [Google Scholar]
- [133].Marracci GH, Jones RE, McKeon GP, Bourdette DN, Alpha lipoic acid inhibits T cell migration into the spinal cord and suppresses and treats experimental autoimmune encephalomyelitis, J. Neuroimmunol 131 (2002) 104–114. [DOI] [PubMed] [Google Scholar]
- [134].Spain R, Powers K, Murchison C, Heriza E, Winges K, Yadav V, et al. , Lipoic acid in secondary progressive MS: a randomized controlled pilot trial, Neurol. Neuroimmunol. Neuroinflamm 4 (2017) e374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Carvalho AN, Lim JL, Nijland PG, Witte ME, van Horssen J, Glutathione in multiple sclerosis: more than just an antioxidant? Mult. Scler 20 (2014) 1425–1431. [DOI] [PubMed] [Google Scholar]
- [136].Morales Pantoja IE, Hu C-L, Perrone-Bizzozero NI, Zheng J, Bizzozero OA, Nrf2-dysregulation correlates with reduced synthesis and low glutathione levels in experimental autoimmune encephalomyelitis, J. Neurochem 139 (2016) 640–650 John Wiley & Sons, Ltd (10.1111). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Stanislaus R, Gilg AG, Singh AK, Singh I, N-acetyl-L-cysteine ameliorates the inflammatory disease process in experimental autoimmune encephalomyelitis in Lewis rats, J. Autoimmune Dis 2 (2005) 4–11 BioMed Central. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Ljubisavljevic S, Stojanovic I, Pavlovic D, Sokolovic D, Stevanovic I, Amino guanidine and N-acetyl-cysteine supress oxidative and nitrosative stress in EAE rat brains, Redox Rep 16 (2011) 166–172. Taylor & Francis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Zhang H, Forman HJ, Choi J, Gamma-glutamyl transpeptidase in glutathione biosynthesis, Meth. Enzymol 401 (2005) 468–483 Elsevier. [DOI] [PubMed] [Google Scholar]
- [140].Slavin J, Fiber and prebiotics: mechanisms and health benefits, Nutrients 5 (2013) 1417–1435 Multidisciplinary Digital Publishing Institute. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Maslowski KM, Mackay CR, Diet, gut microbiota and immune responses, Nat. Publ. Group 12 (2011) 5–9. [DOI] [PubMed] [Google Scholar]
- [142].Koh A, De Vadder F, Kovatcheva-Datchary P, Backhed F, From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites, Cell 165 (2016) 1332–1345. [DOI] [PubMed] [Google Scholar]
- [143].Berer K, Martinez I, Walker A, Kunkel B, Schmitt-Kopplin P, Walter J, et al. , Dietary non-fermentable fiber prevents autoimmune neurological disease by changing gut metabolic and immune status, Sci. Rep 8 (2018) Nature Publishing Group. (10431–12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Mizuno M, Noto D, Kaga N, Chiba A, Miyake S, The dual role of short fatty acid chains in the pathogenesis of autoimmune disease models. Ashour HM, editor, PLoS One 12 (2017) Public Library of Science. (eOl73032). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Rooks MG, Garrett WS, Gut microbiota, metabolites and host immunity, Nat. Rev. Immunol 16 (2016) 341–352 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Usami M, Kishimoto K, Ohata A, Miyoshi M, Aoyama M, Fueda Y, et al. , Butyrate and trichostatin a attenuate nuclear factor kappaB activation and tumor necrosis factor alpha secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells, Nutr. Res 28 (2008) 321–328. [DOI] [PubMed] [Google Scholar]
- [147].Chang PV, Hao L, Offermanns S, Medzhitov R, The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition, Proc. Natl. Acad. Sci. USA 111 (2014) 2247–2252 National Academy of Sciences. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [148].Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, et al. , Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis, Nat. Med 20 (2014) 159–166 Nature Publishing [DOI] [PubMed] [Google Scholar]
- [149].Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, et al. , The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis, Science 341 (2013) 569–573. American Association for the Advancement of Science. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation, Nature 504 (2013) 451–455 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. , Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells, Nature 504 (2013) 446–450 Nature Publishing Group. [DOI] [PubMed] [Google Scholar]
- [152].Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, et al. , Host microbiota constantly control maturation and function of microglia in the CNS, Nat. Neurosci 18 (2015) 965–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Tóth M, et al. , The gut microbiota influences blood-brain barrier permeability in mice, Sci. Transl. Med 6 (2014) (263ral58–8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Luu M, Pautz S, Kohl V, Singh R, Romero R, Lucas S, et al. , The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes, Nat. Commun 10 (2019) Nature Publishing Group. (760–12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Chen T, Noto D, Hoshino Y, Mizuno M, Miyake S, Butyrate suppresses demyelination and enhances remyelination, J. Neuroinflammation 16 (2019) BioMed Central. (165–13). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Park J, Wang Q, Wu Q, Mao-Draayer Y, Kim CH, Bidirectional regulatory potentials of short-chain fatty acids and their G-protein-coupled receptors in autoimmune neuroinflammation, Sci Rep 9 (2019) Nature Publishing Group. (8837–13). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Duscha A, Gisevius B, Hirschberg S, Yissachar N, Stangl GI, Eilers E, et al. , Propionic Acid Shapes the Multiple Sclerosis Disease Course by an Immunomodulatory Mechanism, Cell 180 (2020) 1067–1080 el 6. [DOI] [PubMed] [Google Scholar]
- [158].Hoppenbrouwers T, Cvejić Hogervorst JH, Garssen J, Wichers HJ, Willemsen LEM, Long chain polyunsaturated fatty acids (LCPUFAs) in the prevention of food allergy, Front. Immunol 10 (2019) 1118 Frontiers. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Yashodhara BM, Umakanth S, Pappachan JM, Bhat SK, Kamath R, Choo BH, Omega-3 fatty acids: a comprehensive review of their role in health and disease, Postgrad. Med. J 85 (2009) 84–90. [DOI] [PubMed] [Google Scholar]
- [160].Simopoulos AP, An increase in the omega-6/omega-3 fatty acid ratio increases the risk for obesity, Nutrients 8 (2016) 128 Multidisciplinary Digital Publishing Institute. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [161].Davies P, Bailey PJ, Goldenberg MM, Ford-Hutchinson AW, The role of arachidonic acid oxygenation products in pain and inflammation, Annu. Rev. Immunol 2 (1984) 335–357. [DOI] [PubMed] [Google Scholar]
- [162].Calder PC, Grimble RF, Polyunsaturated fatty acids, inflammation and immunity, Eur. J. Clin. Nutr 56 (Suppl. 3) (2002) S14–S19 Nature Publishing Group. [DOI] [PubMed] [Google Scholar]
- [163].Sperling RI, L Benincaso A, Knoell CT, Larkin JK, Austen KF, Robinson DR, Dietary omega-3 polyunsaturated fatty acids inhibit phosphoinositide formation and chemotaxis in neutrophils, J. Clin. Invest 91 (1993) 651–660. American Society for Clinical Investigation. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Patterson E, Wall R, Fitzgerald GF, Ross RP, Stanton C, Health implications of high dietary omega-6 polyunsaturated fatty acids, J. Nutr. Metab 2012 (2012) Hindawi. (539426–16). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [165].Kouchaki E, Afarini M, Abolhassani J, Mirhosseini N, Bahmani F, Masoud SA, et al. , High-dose o>3 fatty acid plus vitamin D3 supplementation affects clinical symptoms and metabolic status of patients with multiple sclerosis: a randomized controlled clinical trial, J. Nutr 148 (2018) 1380–1386. [DOI] [PubMed] [Google Scholar]
- [166].Langer-Gould A, Black LJ, Waubant E, Smith JB, Wu J, Gonzales EG, et al. , Seafood, fatty acid biosynthesis genes, and multiple sclerosis susceptibility, Mult Scler, vol. 155, SAGE Publications, London, England, 2019(135245851987265). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [167].Torkildsen Ø, Wergeland S, Bakke S, Beiske AG, Bjerve KS, Hovdal H, et al. , ω-3 fatty acid treatment in multiple sclerosis (OFAMS Study): a randomized, double-blind, placebo-controlled trial, Arch. Neurol 69 (2012) 1044–1051. [DOI] [PubMed] [Google Scholar]
- [168].Ascherio A, Munger KL, Environmental risk factors for multiple sclerosis. Part I: the role of infection, Ann. Neurol 61 (2007) 288–299. [DOI] [PubMed] [Google Scholar]
- [169].Goldacre MJ, Wotton CJ, Seagroatt V, Yeates D, Multiple sclerosis after in-fectious mononucleosis: record linkage study, J. Epidemiol. Community Health 58 (2004) 1032–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Sumaya CV, Myers LW, Ellison GW, Epstein-Barr virus antibodies in multiple sclerosis, Arch. Neurol 37 (1980) 94–96. [DOI] [PubMed] [Google Scholar]
- [171].Blackmore S, Hernandez J, Juda M, Ryder E, Freund GG, Johnson RW, et al. , Influenza infection triggers disease in a genetic model of experimental autoimmune encephalomyelitis, Proc. Natl. Acad. Sci. U. S. A 114 (2017) E6107–E6116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [172].Kriesel JD, Bhetariya P, Wang Z-M, Renner D, Palmer C, Fischer KF, Spectrum of microbial sequences and a bacterial cell wall antigen in primary demyelination brain specimens obtained from living patients, Sci. Rep 9 (2019) 1387 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [173].Lee YK, Menezes JS, Umesaki Y, Mazmanian SK, Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis, Proc. Natl. Acad. Sci. USA 108 (Suppl. 1) (2011) 4615–4622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Berer K, Mues M, Koutrolos M, Rasbi ZA, Boziki M, Johner C, et al. , Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination, Nature 479 (2011) 538–541 Nature Publishing Group. [DOI] [PubMed] [Google Scholar]
- [175].Yokote H, Miyake S, Croxford JL, Oki S, Mizusawa H, Yamamura T, NKT cell-dependent amelioration of a mouse model of multiple sclerosis by altering gut flora, Am. J. Pathol 173 (2008) 1714–1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Ochoa-Repáraz J, Mielcarz DW, Ditrio LE, Burroughs AR, Foureau DM, Haque-Begum S, et al. , Role of gut commensal microflora in the development of experimental autoimmune encephalomyelitis, J. Immunol 183 (2009) 6041–6050. [DOI] [PubMed] [Google Scholar]
- [177].Ochoa-Reparaz J, Mielcarz DW, Ditrio LE, Burroughs AR, Begum-Haque S, Dasgupta S, et al. , Central nervous system demyelinating disease protection by the human commensal Bacteroides fragilis depends on polysaccharide a expression, J. Immunol 185 (2010) 4101–4108. [DOI] [PubMed] [Google Scholar]
- [178].He B, Hoang TK, Tian X, Taylor GM, Blanchard E, Luo M, et al. , Lactobacillus reuteri reduces the severity of experimental autoimmune encephalomyelitis in mice by modulating gut microbiota, Front. Immunol 10 (2019) 385 Frontiers. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [179].Lavasani S, Dzhambazov B, Nouri M, Fåk F, Buske S, Molin G, et al. , A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells, PLoS One 5 (2010) e9009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Maassen CBM, Claassen E, Strain-dependent effects of probiotic lactobacilli on EAE autoimmunity, Vaccine 26 (2008) 2056–2057. [DOI] [PubMed] [Google Scholar]
- [181].Libbey JE, Sanchez JM, Doty DJ, Sim JT, Cusick MF, Cox JE, et al. , Variations in diet cause alterations in microbiota and metabolites that follow changes in disease severity in a multiple sclerosis model, Benef. Microbes 9 (2018) 495–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Salehipour Z, Haghmorad D, Sankian M, Rastin M, Nosratabadi R, Soltan Dallal MM, et al. , Bifidobacterium animalis in combination with human origin of lactobacillus plantarum ameliorate neuroinflammation in experimental model of multiple sclerosis by altering CD4 + T cell subset balance, Biomed. Pharmacother 95 (2017) 1535–1548. [DOI] [PubMed] [Google Scholar]
- [183].Mangalam A, Shahi SK, Luckey D, Karau M, Marietta E, Luo N, et al. , Human gut-derived commensal bacteria suppress CNS inflammatory and demyelinating disease, CellReports 20 (2017) 1269–1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Miyake S, Kim S, Suda W, Oshima K, Nakamura M, Matsuoka T, et al. , Dysbiosis in the gut microbiota of patients with multiple sclerosis, with a striking depletion of species belonging to Clostridia XlVa and IV clusters. Wilson BA, editor, PLoS One 10 (2015) Public Library of Science. (eOl 37429). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [185].Chen J, Chia N, Kalari KR, Yao JZ, Novotna M, Soldan MMP, et al. , Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls, Nat. Publ. Group (2016) 1–10 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Jangi S, Gandhi R, Cox LM, Li N, von Glehn F, Yan R, et al. , Alterations of the human gut microbiome in multiple sclerosis, Nat. Comm 7 (2016) 12015 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [187].Cekanaviciute E, Yoo BB, Runia TF, Debelius JW, Singh S, Nelson CA, et al. , Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models, Proc. Natl. Acad. Sci. U. S. A 114 (2017) 10713–10718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Berer K, Gerdes LA, Cekanaviciute E, Jia X, Xiao L, Xia Z, et al. , Gut microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalomyelitis in mice, Proc. Natl. Acad. Sci. U. S. A 114 (2017) 10719–10724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Nguyen N-P, Warnow T, Pop M, White B, A perspective on 16S rRNA operational taxonomic unit clustering using sequence similarity, NPJ Biofilms Microbiomes 2 (2016) 16004–16008 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. , Human gut microbiome viewed across age and geography, Nature 486 (2012) 222–227 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [191].Katz Sand I, Zhu Y, Ntranos A, Clemente JC, Cekanaviciute E, Brandstadter R, et al. , Disease-modifying therapies alter gut microbial composition in MS, Neurol. Neuroimmunol. Neuroinflamm 6 (2019) e517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [192].D’Amore R, Ijaz UZ, Schirmer M, Kenny JC, Gregory R, Darby AC, et al. , A comprehensive benchmarking study of protocols and sequencing platforms for 16S rRNA community profiling, BMC Genomics 17 (2016) BioMed Central. (55–20). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Pollock J, Glendinning L, Wisedchanwet T, Watson M, The madness of micro-biome: attempting to find consensus “best practice” for 16S microbiome studies. S-J Liu, editor, Appl. Environ. Microbiol 84 (2018) 3225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [194].Tamtaji OR, Kouchaki E, Salami M, Aghadavod E, Akbari E, Tajabadi-Ebrahimi M, et al. , The effects of probiotic supplementation on gene expression related to inflammation, insulin, and lipids in patients with multiple sclerosis: a randomized, double-blind, placebo-controlled trial, J. Am. Coll. Nutr 36 (2017) 660–665. Taylor & Francis. [DOI] [PubMed] [Google Scholar]
- [195].Kouchaki E, Tamtaji OR, Salami M, Bahmani F, Daneshvar Kakhaki R, Akbari E, et al. , Clinical and metabolic response to probiotic supplementation in patients with multiple sclerosis: a randomized, double-blind, placebo-controlled trial, Clin. Nutr 36 (2017) 1245–1249. [DOI] [PubMed] [Google Scholar]
- [196].Tankou SK, Regev K, Healy BC, Cox LM, Tjon E, Kivisäkk P, et al. , Investigation of probiotics in multiple sclerosis, Mult Scler, vol. 24, SAGE Publications, London, England, 2018, pp. 58–63. [DOI] [PubMed] [Google Scholar]
- [197].Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, et al. , Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity, Science vol. 352, (2016) 565–569. American Association for the Advancement of Science. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [198].Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, et al. , Population-level analysis of gut microbiome variation, Science vol. 352, (2016) 560–564. American Association for the Advancement of Science. [DOI] [PubMed] [Google Scholar]
- [199].Kolodziejczyk AA, Zheng D, Elinav E, Diet-micro biota interactions and personalized nutrition, Nat. Rev. Micro 13 (2019) Nature Publishing Group. (260–12). [DOI] [PubMed] [Google Scholar]
- [200].Zmora N, Suez J, Elinav E, You are what you eat: diet, health and the gut microbiota, Nat. Rev. Gastroenterol. Hepatol 16 (2019) 35–56 Nature Publishing Group. [DOI] [PubMed] [Google Scholar]
- [201].Koropatkin NM, Cameron EA, Martens EC, How glycan metabolism shapes the human gut microbiota, Nat Rev. Micro 10 (2012) 323–335 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [202].Cowan MM, Plant products as antimicrobial agents, Clin. Microbiol. Rev 12 (1999) 564–582. American Society for Microbiology (ASM). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [203].Li Y, Innocentin S, Withers DR, Roberts NA, Gallagher AR, Grigorieva EF, et al. , Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation, Cell 147 (2011) 629–640. [DOI] [PubMed] [Google Scholar]
- [204].Ooi JH, Li Y, Rogers CJ, Cantorna MT, Vitamin D regulates the gut microbiome and protects mice from dextran sodium sulfate-induced colitis, J. Nutr 143 (2013) 1679–1686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [205].Kaliannan K, Wang B, Li X-Y, Kim K-J, Kang JX, A host-microbiome interaction mediates the opposing effects of omega-6 and omega-3 fatty acids on metabolic endotoxemia, Sci. Rep 5 (2015) 11276 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [206].Su D, Nie Y, Zhu A, Chen Z, Wu P, Zhang L, et al. , Vitamin D signaling through induction of paneth cell defensins maintains gut microbiota and improves metabolic disorders and hepatic steatosis in animal models, Front. Physiol 7 (2016) 498 Frontiers. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [207].Hibberd MC, Wu M, Rodionov DA, Li X, Cheng J, Griffin NW, et al. , The effects of micronutrient deficiencies on bacterial species from the human gut microbiota, Sci. Transl. Med 9 (2017) American Association for the Advancement of Science. (eaal4069). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [208].Chassaing B, Koren O, Goodrich JK, Poole AC, Srinivasan S, Ley RE, et al. , Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome, Nature 519 (2015) 92–96 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [209].David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. , Diet rapidly and reproducibly alters the human gut microbiome, Nature 505 (2014) 559–563 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [210].Shulman RJ, Hollister EB, Cain K, Czyzewski DI, Self MM, Weidler EM, et al. , Psyllium fiber reduces abdominal pain in children with irritable bowel syndrome in a randomized, Double-Blind Trial, Clin. Gastroenterol. Hepatol 15 (2017) 712–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [211].Korem T, Zeevi D, Zmora N, Weissbrod O, Bar N, Lotan-Pompan M, et al. , Bread affects clinical parameters and induces gut microbiome-associated personal glycemic responses, Cell Metab 25 (2017) 1243–1245. [DOI] [PubMed] [Google Scholar]
- [212].Wu GD, Chen J, Hoffmann C, Bittinger K, Chen Y-Y, Keilbaugh SA, et al. , Linking long-term dietary patterns with gut microbial enterotypes, Science 334 (2011) 105–108. American Association for the Advancement of Science. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [213].Quintana FJ, Sherr DH. Aryl hydrocarbon receptor control of adaptive immunity. Insel PA, editor, Pharmacol. Rev 65 (2013) 1148–1161. American Society for Pharmacology and Experimental Therapeutics. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [214].Rothhammer V, Mascanfroni ID, Bunse L, Takenaka MC, Kenison JE, Mayo L, et al. , Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor, Nat. Med 22 (2016) 586–597 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [215].Nourbakhsh B, Bhargava P, Tremlett H, Hart J, Graves J, Waubant E, Altered tryptophan metabolism is associated with pediatric multiple sclerosis risk and course, Ann. Clin. Transl. Neurol 5 (2018) 1211–1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [216].Libbey JE, Doty DJ, Sim JT, Cusick MF, Round JL, Fujinami RS, The effects of diet on the severity of central nervous system disease: one part of lab-to-lab variability, Nutrition 32 (2016) 877–883 Elsevier Inc. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [217].Tremlett H, Fadrosh DW, Faruqi AA, Zhu F, Hart J, Roalstad S, et al. , Gut microbiota in early pediatric multiple sclerosis: a case-control study, Eur. J. Neurol 23 (2016) 1308–1321 John Wiley & Sons, Ltd (10.1111). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [218].Hughes LE, Smith PA, Bonell S, Natt RS, Wilson G, Rashid T, et al. , Cross-reactivity between related sequences found in Acinetobacter sp., Pseudomonas aeruginosa, myelin basic protein and myelin oligodendrocyte glycoprotein in multiple sclerosis, J. Neuroimmunol 144 (2003) 105–115. [DOI] [PubMed] [Google Scholar]
- [219].JO-RA Raz, Mielcarz DW, Wang Y, Begum-Haque S, Dasgupta S, Kasper DL, et al. , A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease, Mucosal Immunol. 3 (2010) 487–495 Nature Publishing Group [DOI] [PubMed] [Google Scholar]
- [220].Wang Y, Telesford KM, Ochoa-Reparaz J, Haque-Begum S, Christy M, Kasper EJ, et al. , An intestinal commensal symbiosis factor controls neuroinflammation via TLR2~mediated CD39 signalling, Nat. Comm 5 (2014) 4432 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [221].Consonni A, Cordiglieri C, Rinaldi E, Marolda R, Ravanelli I, Guidesi E, et al. , Administration of bifidobacterium and lactobacillus strains modulates experimental myasthenia gravis and experimental encephalomyelitis in Lewis rats, Oncotarget 9 (2018) 22269–22287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [222].Omenetti S, Bussi C, Metidji A, Iseppon A, Lee S, Tolaini M, et al. , The intestine harbors functionally distinct homeostatic tissue-resident and inflammatory Th17 cells, Immunity 51 (2019) (77–89. e6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [223].Chen H, Ma X, Liu Y, Ma L, Chen Z, Lin X, et al. , Gut microbiota interventions with Clostridium butyricum and norfloxacin modulate immune response in experimental autoimmune encephalomyelitis mice, Front. Immunol 10 (2019) 1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [224].Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. , Induction of colonic regulatory T cells by indigenous Clostridium species, Science 331 (2011) 337–341. American Association for the Advancement of Science. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [225].Zhou L, Zhang M, Wang Y, Dorfman RG, Liu H, Yu T, et al. , Faecalibacterium prausnitzii produces butyrate to maintain Th17/Treg balance and to ameliorate colorectal colitis by inhibiting histone deacetylase 1, Inflamm. Bowel Dis 24 (2018) 1926–1940. [DOI] [PubMed] [Google Scholar]
- [226].Ye P, Garvey PB, Zhang P, Nelson S, Bagby G, Summer WR, et al. , Interleukin-17 and lung host defense against Klebsiella pneumoniae infection, Am. J. Respir. Cell Mol. Biol 25 (2001) 335–340. American Thoracic Society, New York, NY. [DOI] [PubMed] [Google Scholar]
- [227].Yamashita M, Ukibe K, Matsubara Y, Hosoya T, Sakai F, Kon S, et al. , Lactobacillus helveticus SBT2171 attenuates experimental autoimmune encephalomyelitis in mice, Front. Microbiol 8 (2017) 2596 Frontiers. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [228].Shapira L, Ayalon S, Brenner T, Effects of Porphyromonas gingivalis on the central nervous system: activation of glial cells and exacerbation of experimental autoimmune encephalomyelitis, J. Periodontol 73 (2002) 511–516. [DOI] [PubMed] [Google Scholar]
- [229].Shahi SK, Freedman SN, Murra AC, Zarei K, Sompallae R, Gibson-Corley KN, et al. , Prevotella histicola, a human gut commensal, is as potent as COPAXONE® in an animal model of multiple sclerosis, Front. Immunol 10 (2019) 462 Frontiers. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [230].Henke MT, Kenny DJ, Cassilly CD, Vlamakis H, Xavier RJ, Clardy J, Ruminococcus gnavus, a member of the human gut microbiome associated with Crohn’s disease, produces an inflammatory polysaccharide, Proc. Natl. Acad. Sci. USA 116 (2019) 12672–12677 National Academy of Sciences. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [231].Herrmann I, Kellert M, Schmidt H, Mildner A, Hanisch UK, Brück W, et al. , Streptococcus pneumoniae infection aggravates experimental autoimmune encephalomyelitis via toll-like receptor 2, Infect. Immun 74 (2006) 4841–4848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [232].Santacruz A, Marcos A, Wärnberg J, Martí A, Martin-Matillas M, Campoy C, et al. , Interplay between weight loss and gut microbiota composition in over-weight adolescents, Obesity (Silver Spring) 17 (2009) 1906–1915 John Wiley & Sons, Ltd. [DOI] [PubMed] [Google Scholar]
- [233].Nadal I, Santacruz A, Marcos A, Warnberg J, Garagorri JM, Garagorri M, et al. , Shifts in Clostridia, bacteroides and immunoglobulin-coating fecal bacteria associated with weight loss in obese adolescents, Int. J. Obes. (Lond.) 33 (2009) 758–767 Nature Publishing Group. [DOI] [PubMed] [Google Scholar]
- [234].Murtaza N, Burke LM, Vlahovich N, Char lesson B, O’Neill H, Ross ML, et al. , The effects of dietary pattern during intensified training on stool microbiota of elite race walkers, Nutrients 11 (2019) 261 Multidisciplinary Digital Publishing Institute. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [235].Zhang Y, Zhou S, Zhou Y, Yu L, Zhang L, Wang Y, Altered gut microbiome composition in children with refractory epilepsy after ketogenic diet, Epilepsy Res 145 (2018) 163–168. [DOI] [PubMed] [Google Scholar]
- [236].Xie G, Zhou Q, Qiu C-Z, Dai W-K, Wang H-P, Li Y-H, et al. , Ketogenic diet poses a significant effect on imbalanced gut microbiota in infants with refractory epilepsy, World J. Gastroenterol 23 (2017) 6164–6171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [237].Tagliabue A, Ferraris C, Uggeri F, Trentani C, Bertoli S, de Giorgis V, et al. , Short-term impact of a classical ketogenic diet on gut microbiota in GLUT1 deficiency syndrome: a 3-month prospective observational study, Clin. Nutr 17 (2017) 33–37 ESPEN. [DOI] [PubMed] [Google Scholar]
- [238].Lindefeldt M, Eng A, Darban H, Bjerkner A, Zetterstrom CK, Allander T, et al. , The ketogenic diet influences taxonomic and functional composition of the gut micro biota in children with severe epilepsy, NPJ Biofilms Microbiomes 5 (2019) 5–13 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [239].Sonnenburg ED, Sonnenburg JL, The ancestral and industrialized gut microbiota and implications for human health, Nat. Rev. Micro 17 (2019) 383–390 Nature Publishing Group. [DOI] [PubMed] [Google Scholar]
- [240].Lin A, Bik EM, Costello EK, Dethlefsen L, Haque R, Reiman DA, et al. , Distinct distal gut microbiome diversity and composition in healthy children from Bangladesh and the United States. Aziz RK, editor, PLoS One 8 (2013) Public Library of Science. (e53838). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [241].Vangay P, Johnson AJ, Ward TL, Al-Ghalith GA, Shields-Cutler RR, Hillmann BM, et al. , US Immigration Westernizes the Human Gut Microbiome, Cell 175 (2018) (962–972. el0). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [242].De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, et al. , Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa, Proc. Natl. Acad. Sci. USA 107 (2010) 14691–14696 National Academy of Sciences. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [243].Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. , A metagenome-wide association study of gut microbiota in type 2 diabetes, Nature 490 (2012) 55–60 Nature Publishing Group. [DOI] [PubMed] [Google Scholar]
- [244].Menni G, Jackson MA, Pallister T, Steves CJ, Spector TD, Valdes AM, Gut microbiome diversity and high-fibre intake are related to lower long-term weight gain, Int. J. Obes. (Lond.) 41 (2017) 1099–1105 Nature Publishing Group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [245].Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. , Richness of human gut microbiome correlates with metabolic markers, Nature 500 (2013) 541–546 Nature Publishing Group. [DOI] [PubMed] [Google Scholar]
- [246].Haro C, García-Carpintero S, Rangel-Zúñiga OA, Alcalá-Díaz JF, Landa BB, Clemente JC, et al. , Consumption of two healthy dietary patterns restored microbiota dysbiosis in obese patients with metabolic dysfunction, Mol. Nutr. Food Res 61 (2017) 1700300. [DOI] [PubMed] [Google Scholar]
- [247].Matijašić BB, Obermajer T, Lipoglavšek L, Grabnar I, Avguštin G, Rogelj I, Association of dietary type with fecal microbiota in vegetarians and omnivores in Slovenia, Eur. J. Nutr 53 (2014) 1051–1064 Springer Berlin Heidelberg. [DOI] [PubMed] [Google Scholar]
- [248].Barrett HL, Gomez-Arango LF, Wilkinson SA, McIntyre HD, Callaway LK, Morrison M, et al. , A vegetarian diet is a major determinant of gut microbiota composition in early pregnancy, Nutrients 10 (2018) 890 Multidisciplinary Digital Publishing Institute. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [249].Ruengsomwong S, La-Ongkham O, Jiang J, Wannissorn B, Nakayama J, Nitisinprasert S, Microbial community of healthy thai vegetarians and non-vegetarians, their core gut microbiota, and pathogen risk, J. Microbiol. Biotechnol 26 (2016) 1723–1735. [DOI] [PubMed] [Google Scholar]
- [250].De Filippis F, Pellegrini N, Vannini L, Jeffery IB, La Storia A, Laghi L, et al. , High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome, Gut 65 (2016) 1812–1821 BMJ Publishing Group. [DOI] [PubMed] [Google Scholar]
- [251].Zimmer J, Lange B, Frick J-S, Sauer H, Zimmermann K, Schwiertz A, et al. , A vegan or vegetarian diet substantially alters the human colonic faecal microbiota, Eur. J. Clin. Nutr 66 (2012) 53–60 Nature Publishing Group. [DOI] [PubMed] [Google Scholar]
- [252].Garcia-Mantrana I, Selma-Royo M, Alcantara C, Collado MC, Shifts on gut microbiota associated to mediterranean diet adherence and specific dietary intakes on general adult population, Front Microbiol 9 (2018) 890 Frontiers. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [253].Pagliai G, Russo E, Niccolai E, Dinu M, Di Pilato V, Magrini A, et al. , Influence of a 3-month low-calorie Mediterranean diet compared to the vegetarian diet on human gut micro biota and SCFA: the CARDIVEG Study, Eur. J. Nutr 11 (2019) Springer Berlin Heidelberg. (227–14). [DOI] [PubMed] [Google Scholar]
- [254].Buchman M, Kruse C, Broach W, Jensen E, Kales S, Composition of Human Gut Microbiota After a Mediterranean Diet Intervention Among Fire Fighters (OR23–05-19), (2019). [Google Scholar]
- [255].Haro C, Montes-Borrego M, Rangel-Zúñiga OA, Alcalá-Díaz JF, Gómez-Delgado F, Pérez-Martínez P, et al. , Two healthy diets modulate gut microbial community improving insulin sensitivity in a human obese population, J. Clin. Endocrinol. Metab 101 (2016) 233–242. [DOI] [PubMed] [Google Scholar]
- [256].Mitsou EK, Kakali A, Antonopoulou S, Mountzouris KC, Yannakoulia M, Panagiotakos DB, et al. , Adherence to the Mediterranean diet is associated with the gut microbiota pattern and gastrointestinal characteristics in an adult population, Br. J. Nutr 117 (2017) 1645–1655 Cambridge University Press. [DOI] [PubMed] [Google Scholar]
- [257].Talsness CE, Penders J, Jansen EHJM, Damoiseaux J, Thijs C, Mommers M, Influence of vitamin D on key bacterial taxa in infant microbiota in the KOALA Birth Cohort Study. Taneja V, editor, PLoS One 12 (2017) Public Library of Science. (eOl 88011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [258].Garg M, Hendy P, Ding JN, Shaw S, Hold G, Hart A, The effect of vitamin D on intestinal inflammation and faecal microbiota in patients with ulcerative colitis, J. Crohns Colitis 12 (2018) 963–972 2nd ed. [DOI] [PubMed] [Google Scholar]
- [259].Sordillo JE, Zhou Y, McGeachie MJ, Ziniti J, Lange N, Laranjo N, et al. , Factors influencing the infant gut microbiome at age 3–6 months: Findings from the ethnically diverse vitamin D antenatal asthma reduction trial (VDAART), J. Allergy Clin. Immunol 139 (2017) (482–491. e14). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [260].Kanhere M, He J, Chassaing B, Ziegler TR, Alvarez JA, Ivie EA, et al. , Bolus weekly vitamin D3 supplementation impacts gut and airway microbiota in adults with cystic fibrosis: a double-blind, randomized, placebo-controlled clinical trial, J. Clin. Endocrinol. Metab 103 (2018) 564–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [261].Luthold RV, Fernandes GR, Franco-de-Moraes AC, Folchetti LGD, Ferreira SRG, Gut microbiota interactions with the immunomodulatory role of vitamin D in normal individuals, Metab. Clin. Exp 69 (2017) 76–86. [DOI] [PubMed] [Google Scholar]
- [262].Bashir M, Prietl B, Tauschmann M, Mautner SI, Kump PK, Treiber G, et al. , Effects of high doses of vitamin D3 on mucosa-associated gut microbiome vary between regions of the human gastrointestinal tract, Eur. J. Nutr 55 (2016) 1479–1489 8 ed. Springer Berlin Heidelberg. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [263].L Queipo-Ortuño M, Boto-Ordóñez M, Murri M, Gomez-Zumaquero JM, Clemente-Postigo M, Estruch R, et al. , Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers, Am. J. Clin. Nutr 95 (2012) 1323–1334. [DOI] [PubMed] [Google Scholar]
- [264].Cuervo A, He via A, López P, Suárez A, Diaz C, Sánchez B, et al. , Phenolic compounds from red wine and coffee are associated with specific intestinal microorganisms in allergic subjects, Food Funct. 7 (2016) 104–109. The Royal Society of Chemistry [DOI] [PubMed] [Google Scholar]
- [265].Yamakoshi J, Tokutake S, Kikuchi M, Kubota Y, Konishi H, Mitsuoka T, Effect of Proanthocyanidin-rich extract from grape seeds on human Fecal Flora and Fecal odor, Microbial Ecol. Health Dis 13 (2009) 25–31. Taylor & Francis. [Google Scholar]
- [266].Tzounis X, Rodriguez-Mateos A, Vulevic J, Gibson GR, Kwik-Uribe C, Spencer JPE, Prebiotic evaluation of cocoa-derived flavanols in healthy humans by using a randomized, controlled, double-blind, crossover intervention study, Am. J. Clin. Nutr 93 (2011) 62–72. [DOI] [PubMed] [Google Scholar]
- [267].Vendrame S, Guglielmetti S, Riso P, Arioli S, Klimis-Zacas D, Porrini M, Six-week consumption of a wild blueberry powder drink increases bifidobacteria in the human gut, J. Agric. Food Chem 59 (2011) 12815–12820. American Chemical Society. [DOI] [PubMed] [Google Scholar]
- [268].Cuervo A, Valdés L, Salazar N, de los Reyes-Gavilán CG, Ruas-Madiedo P, Gueimonde M, et al. , Pilot study of diet and microbiota: interactive associations of fibers and polyphenols with human intestinal bacteria, J. Agric. Food Chem 62 (2014) 5330–5336. American Chemical Society. [DOI] [PubMed] [Google Scholar]