Next Article in Journal
Profiles of Early Childhood Adversity in an Urban Pediatric Clinic: Implications for Pediatric Primary Care
Next Article in Special Issue
Who Is Best Placed to Support Cyber Responsibilized UK Parents?
Previous Article in Journal
Hazardous Child Labour, Psychosocial Functioning, and School Dropouts among Children in Bangladesh: A Cross-Sectional Analysis of UNICEF’s Multiple Indicator Cluster Surveys (MICS)
Previous Article in Special Issue
Parent Perceptions of an Early Childhood System’s Community Efforts: A Qualitative Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Multiple Sclerosis—Related Dietary and Nutritional Issues: An Updated Scoping Review with a Focus on Pediatrics

1
Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy
2
Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, 80138 Naples, Naples, Italy
3
Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80138 Naples, Naples, Italy
4
Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Nutrition Section, University of Salerno, 84081 Baronissi, Salerno, Italy
5
Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatric Psychiatry Section, University of Salerno, 84081 Baronissi, Salerno, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Children 2023, 10(6), 1022; https://doi.org/10.3390/children10061022
Submission received: 16 May 2023 / Revised: 3 June 2023 / Accepted: 5 June 2023 / Published: 7 June 2023

Abstract

:
Purpose. Lifestyle/dietetic habits play an important role in the development and progression of multiple sclerosis (MS) disease. Here, we examine the basic pathomechanisms underlying intestinal and brain barrier modifications in MS and consider diets and dietary supplementations proposed over time to complement pharmacological therapies for improving disease outcome both in adults and in children. Methods. Scoping literature search about evidence-based findings in MS-related gut-brain axis (GBA) pathophysiology and nutritional issues at all ages. Findings. Data show that (1) no universal best diet exists, (2) healthy/balanced diets are, however, necessary to safeguard the adequate intake of all essential nutrients, (3) diets with high intakes of fruits, vegetables, whole grains, and lean proteins that limit processed foods, sugar, and saturated fat appear beneficial for their antioxidant and anti-inflammatory properties and their ability to shape a gut microbiota that respects the gut and brain barriers, (4) obesity may trigger MS onset and/or its less favorable course, especially in pediatric-onset MS. Vitamin D and polyunsaturated fatty acids are the most studied supplements for reducing MS-associated inflammation. Conclusions. Pending results from other and/or newer approaches targeting the GBA (e.g., pre- and probiotics, engineered probiotics, fecal-microbiota transplantation), accurate counseling in choosing adequate diet and maintaining physical activity remains recommended for MS prevention and management both in adults and children.

1. Introduction

Multiple sclerosis (MS) is a disabling immune-mediated demyelinating neurodegenerative disease with an estimated prevalence of 1 in 1000 in populations of European descent. It primarily affects females (F:M = 2–3:1) mainly between the ages of 15 and 55 years. Pediatric-onset MS (POMS) is less common, and specific data for this age group are limited due to sample size and varying levels of functionality and cognitive maturation [1,2].
The disease results from a complex interplay of immunological, genetic, and environmental factors [3,4] (Figure 1).
Studies in both murine models and patients suggest that an autoimmune CD4+ T cell-mediated inflammatory reaction initiates the demyelination process. These activated T cells, triggered by an unknown antigen, can cross the blood-brain barrier (BBB) and initiate an immune attack on the central nervous system (CNS). Inflammatory mediators released by T cells damage myelin, axons, and oligodendrocytes by recruiting and stimulating other immune cells such as B cells. This cascade leads to the formation of scar tissue (sclerosis) when myelin and nerve fibers are destroyed [5]. Genetic factors, particularly within the major histocompatibility complex (e.g., HLA DRB1*1501), contribute to about 50% of the risk of developing MS, although familial MS accounts for less than 15% of cases [5,6]. Environmental factors such as geographic location, infections (e.g., Epstein–Barr virus), smoking, toxins, nutritional deficiencies (especially vitamin D), sedentary lifestyle, unbalanced diets, changes in the gut microbiome, and pediatric-onset obesity have been implicated as risk factors for MS [7]. Recent evidence suggests a significant increase in the incidence of MS worldwide, particularly in Western countries, highlighting the importance of environmental factors in its pathogenesis [8]. Nutritional and dietary factors have gained increasing attention as potential modifiers of MS pathophysiology, aiming to complement current disease-modifying treatments (DMT) targeting inflammation and immune cells [9,10,11,12].The gut-brain axis (GBA), which is closely linked to nutrition and obesity, is also emerging as a critical player in MS [13]. This review aims to examine in-depth the latest growing literature on nutritional and dietary aspects contributing mechanistically to MS with a focus on their potential role as disease modifiers. Additionally, relevant emerging data pertaining to pediatric age will be reviewed to assess whether, as in adult MS, POMS disease management might benefit from add-on nutritional interventions.

2. Methods

This narrative review considered English language scientific literature articles published in the last 5 years, retrieved from the PubMed/Scopus and Google Scholar databases including observational and interventional studies, as well as both systematic and non-systematic reviews dealing with the subjects of nutrients/nutrition/diet/gut-brain-axis/blood-brain barrier/multiple sclerosis/pediatric-onset multiple sclerosis. Documents released as “grey literature” by the most relevant international health agencies and scientific associations made available on multiple electronic scoping searches as of April 2023 and relevant references quoted in the retrieved articles were also considered.

3. The Gut-Brain Axis in MS

The MS GBA contemplates a dysbiosis-induced pro-inflammatory gut environment responsible for a leaky gut [14] allowing the emergence of activated myelin-specific bystander T cells which regulate the cytokine milieu in the central nervous system (CNS) and the function of neurons and glial cells [13]. Clinical [relapsing-remitting (RR) course with subsequent progressive disability] and pathological similarities between MS and the murine model of experimental autoimmune encephalomyelitis (EAE) allow the latter to be considered as a suitable model for the study of MS. Through this model, studies have confirmed that high-fat diets, especially if high in saturated fats, are relevant MS triggers [15,16]. Obesity itself, due to the adipose tissue‘s inflammatory properties mediated by adipokines production (e.g., adiponectin), has been suggested to contribute to the pro-inflammatory status of MS, which also has impacts on the quiescence of CNS-resident microglia [17,18].

3.1. The Intestinal Barrier

The intestinal barrier is a complex functional unit composed of mucosal and luminal elements (i.e., epithelial cells layer; mucosal barrier; innate and acquired immune components); neuroenteric, vascular, and endocrine systems; digestive enzymes; and gut microbiota (GM). This barrier plays a key role in protecting against enteric organisms, their toxins, and bio-products associated with the health and disease susceptibility of organs/systems [19,20]. The recently discovered gut-vascular barrier controls the translocation of gut bacteria and antigens into the bloodstream [21,22].
The gut-(liver)-brain axis connects the GM, neuroendocrine and neuroimmune systems, autonomic nervous system, and enteric nervous system with the CNS [13,23]. The GM consists of trillions of commensal microorganisms that maintain the integrity of the mucosal barrier and contribute to normal host physiology [19]. Factors such as an unbalanced diet, infections, antibiotics, stress, and environmental factors can lead to dysbiosis and increased intestinal permeability [24,25,26]. Dysbiosis is associated with various diseases, including gastrointestinal and systemic inflammatory diseases [27].
GM dysbiosis can also impact the onset and progression of neurological disorders such as MS by affecting metabolic pathways and interacting with host immunity [28,29,30,31,32].
Identifying a characteristic/diagnostic composition of microbial communities associated with the gut microbiota of MS patients is challenging due to variations between studies and individual profiles [31,32]. However, reduced microbial diversity is commonly observed, which is characterized by an increased Firmicutes/Bacteroides ratio and prevalence of species producing endogenous ethanol, lipopolysaccharide, and reactive oxygen species [33,34].
These biochemical factors, along with pro-inflammatory T-helper types and cytokine patterns, contribute to intestinal inflammation and impairment of the barrier function (leaky gut) [29,35].
In an MS mouse model, Streptococcus thermophilus ST285 has been observed to switch cytokine responses to myelin peptides from pro-inflammatory to anti-inflammatory patterns [36]. The significance of increased abundance of Akkermansia species in MS patients and the EAE mouse model is still debated, with suggestions of a compensatory effect rather than a direct association with EAE progression and MS pathogenesis [37,38].
Considering differences in patient selection methods, environmental factors, and dietary habits, certain species (e.g., Faecalibacterium, Eubacterium rectale, Corynebacterium, Fusobacteria, Bacteroides stercoris, and Bacteroides coprocola) [29,39] are generally reduced in MS patients compared to healthy controls in some studies. These species are responsible for decreased production of metabolites such as bacterial lipid 654, a Toll-like receptor 2 ligand derived from gastrointestinal and oral bacteria [40,41,42], and short-chain fatty acids (SCFAs) such as butyrate and propionic acid [14,43,44].
SCFAs, produced by the colonic fermentation of dietary fibers and resistant starch, are speculated to play a key role in neuro-immunoendocrine regulation [45]. A secondary increase in species related to oxidative levels is observed in progressive MS or more severe disease forms [46,47,48], and it is sufficient to induce EAE sensitivity [49,50]. Saturated fatty acids (FAs) have a close relationship with autoimmunity phenomena in MS, as their concentration and composition regulate immune cell polarization, differentiation, and function, with a protective role in blood-brain barrier function. They are critical players in CNS chronic inflammation, progressive degeneration, and remyelination [34,51].
Additionally, a negative association with polysaccharide-digesting bacteria such as B. thetaiotaomicron is often observed [52].

3.2. The Blood-Brain Barrier

The BBB plays a crucial role in maintaining the homeostasis of the brain microenvironment, and its dysfunction is implicated in the pathogenesis of various neurological diseases, including MS. The BBB consists of multiple biological barriers including the proper blood-brain barrier, the blood-cerebrospinal fluid barrier, and the arachnoid barrier [22,53] (Figure 2).
The integrity of the BBB can be influenced by the GM, as bacterial antigens such as lipopolysaccharide (LPS) and SCFAs can travel from the leaky gut to the brain endothelial cells’ Toll-like receptors 2 through the bloodstream. In the EAE murine model, high-fat-diet-induced obesity resulted in severe disease accompanied by gut dysbiosis, increased gut permeability, and systemic inflammation, suggesting a role for gut barrier modulation in obesity-induced MS severity [39]. In a similar EAE model, obesity induced by a high-fat diet also led to BBB disruption, which allows the infiltration of monocytes/macrophages and activation of resident microglia, ultimately exacerbating CNS inflammation in EAE [54], likely mediated by IL-6 and CCL-2 [55].
The activation of pro-inflammatory pathways disrupts the delicate balance between protective and harmful reactions. It can induce the release of Vascular Endothelial Growth Factor B/VEGF-B and Tumor Growth Factor/TGF-alpha from microglia, which activates astrocytes and exerts detrimental effects on neurons. The Th17 cytokines IL-17A and IL-17F appear to be pivotal in triggering BBB disturbance [56,57]. As mentioned earlier, SCFAs not only have effects on the colon and peripheral tissues but also play a crucial role in the communication between the GM, gut, and brain. They can cross the BBB through endothelial cell monocarboxylate transporters and a) upregulate the expression of tight junction proteins essential for BBB integrity, b) affect neuroinflammation by influencing glial cell morphology and function, and modulating levels of neurotrophic factors [45]. Moreover, due to its bidirectional nature, GBA neuroinflammation could result in further intestinal inflammation as the disease progresses by affecting efferent cholinergic transmission [13,58,59].

4. Diets and Dietary Supplementations

4.1. Dietary Influence on MS

Studies in adults with MS suggest that diet-related inflammation increases the odds of developing the disease [60,61]. The prevalence of Westernized diets, characterized by ultra-processed foods that are high in salt, saturated and trans fatty acids [62] and low in fibers and flavonoids, may contribute to the upregulation of pro-inflammatory compounds, gut dysbiosis, neuroinflammation, and neurodegeneration [63,64,65]. The high salt content in processed foods has been associated with disease exacerbation and the development of new lesions, although the evidence is debated [66,67,68]. Saturated fats activate pro-inflammatory Toll-like receptors (TLRs) and increase NF-κB, which affects the innate immune system [7]. They also play a role in GM-mediated inflammation in MS development and relapse risk, as seen in the EAE model [39,54,55] or in MS patients. [63,65,69].
Conversely, higher fish consumption, particularly oily fish rich in vitamin D and omega-3 fatty acids, is associated with a lower risk of CNS demyelination [70]. Flavonoids, polyphenolic compounds that are abundant in fruits and vegetables, have shown a protective role in MS development in experimental models [71,72]. Non-fermentable cellulose fiber may prevent changes in GM composition and T cell responses associated with CNS autoimmunity in MS [50]. The proper functioning of all players in the gut-brain axis is crucial for managing the impact of MS [Table 1].
Several diets have been proposed for MS based on assumptions such as existing food allergies, gluten sensitivity, hypovitaminosis, or the concept of a healthy diet (e.g., Mediterranean diets, Paleolithic diet) [73]. Although a diet rich in fruits and vegetables seems logically protective against relapses and disease progression [74], larger and better-conducted studies are needed to confirm this correlation due to conflicting evidence [75].
Table 1. Synopsis of the main specific diets proposed over time in patients with multiple sclerosis.
Table 1. Synopsis of the main specific diets proposed over time in patients with multiple sclerosis.
Diet Name Main Characteristics
Allergen free/milk free Hypoallergenic diet based on the unproven hypothesis of the association between MS and external allergens [76]. The milk protein butyrophilin has been implicated through antigenic mimicry with myelin oligodendrocyte glycoprotein in EAE [77] as well as in MS patients [78]. Some studies with questionnaires suggest an inverse relationship between total dairy intake and MS disability severity [79,80] with an inverse relationship between whole grain intake and MS-related disability [80].
Gluten freeAmong studies, only one clinical trial gave meaningful results, but there are methodological limitations [81,82,83]. All in all, the current level of evidence is inadequate to state whether gluten plays a role in MS [82].
Mega AscorbicHigh in vitamin C diet. No well-defined link between MS and vitamin C [84].
Multi Vitaminic Multi vitaminic supplementation (e.g., A and D): quite convincing data show that higher vitamin intake/serum levels correlate with lower risk of MS development but not convincing on the contrary [85]. Possible detrimental effects of overdosing require vitamin-level monitoring [86,87].
Hebener’sSelf-reported disease stability/amelioration in one study with fish oil and antioxidant drugs supplementation + Ω-6 restriction [88].
KousmineHigh in polyunsaturated fats/low in animal fats diet to counteract a possibly increased membrane permeability [89,90,91].
Swank (low saturated fats)Low-saturated fats (<20 g fat/day or <20% total calories): reported lower death rates and better outcome in the more adherent patients and those with lower disability at entry [92,93].
Mediterranean diets (MD)Common features include emphasis on vegetables, fruits, beans, nuts, seeds, breads, unrefined grains, and olive oil; inclusion of fish and wine; minimal intake of full-fat dairy products and possibly lean meats [94,95]. Conflicting results on whether lean and unprocessed red meat is detrimental [96,97,98]. It is considered beneficial for its antioxidant properties. Negatively associated with neurological and fatigue symptoms.
Adherence should be monitored through validated tests [e.g., Predimed for adults [94] and KidMed for children [99]
Mediterranean/DASH It derives from the Mediterranean Dietary Approaches to Stop Hypertension (DASH) [100,101].
MINDThe Mediterranean/Intervention for Neurodegenerative Delay (MIND) is a combination of MD and DASH [100,101,102].
Paleolithic1 Consists of high-quality foods full of nutrients and fiber and with less artificial sugar and salt compared to present-day diets [103]. Nutrients included in this diet are essential to myelin growth and repair. Typically, it does not permit consumption of dairy or grain products.
Modified Paleolithic
(MD-PI intervention)
This diet is rich in α-lipoic acid and polyphenols. It has commonalities with MD including avoidance of high-fat meats/ultra-processed foods with added sugar, sodium, and hydrogenated fats [104,105,106].
Wahls™ Paleo dietDifferences from a traditional Paleo diet: exclusion of eggs; limited animal and fish protein. It allows legumes (e.g., soy milk), two servings of gluten-free grains (e.g., rice) per week; it specifies nine cups of fruits and vegetables (F/V)/day with 1/3 each from dark-green leafy vegetables, sulfur-rich vegetables, and deeply colored F/V; seaweed, algae and nutritional yeast are encouraged [107,108].
Wahls/Elim Paleo This is a paleo version modified by adding a restriction of lectins to reduce intestinal permeability and CNS inflammation [107,108].
Overcoming MS (OMS)Minimized saturated fats and plant-based, whole-food diet plus seafood [79,109].
Ketogenic diet (KD)Eliminating all/almost all carbohydrates and increasing the intake of proteins. KD combined with a modified MD have been suggested to improve neuroinflammation in MS [110,111].
Energy restriction (ER) Chronic ER/Intermittent energy restriction (IER) determines a switch from glucose to fatty acids and ketones as the major fuel source for cells [112,113,114,115].
Mice fed a “fasting mimicking” diet (very low-calorie diet lasting for 3 days every 7 days) exhibited delayed onset, reduced incidence, and decreased severity of EAE. Histological findings show reduced immune cell infiltration and demyelination in the spinal cord [116].
McDougall DietA low-fat (10–15% of calories from fat), starch-based, vegan diet with no oils permitted. For 7 days, produced significant favorable changes in commonly tested biomarkers used to predict future risks for cardiovascular disease and metabolic diseases [75,117].
It appeared safe and effective in preventing clinical attacks/new MRI lesions. Drawback = long-term adherence [118].
Abbreviations: DASH, Dietary Approaches to Stop Hypertension; ER, Energy restriction; F/V, fruit and vegetables; KD, Ketogenic diet; MD, Mediterranean diet; MIND, Mediterranean/Intervention for Neurodegenerative Delay; MS, multiple sclerosis; OMS Overcoming MS; PI, Paleolithic intervention.
A recent meta-analysis [119,120] examined 11 studies (involving 608 participants) that investigated whole dietary approaches without concomitant interventions with appropriate control groups and outcome measurements. The modified Paleolithic and modified Mediterranean diets improved fatigue and quality of life compared to the control diets. Low-fat diets improved fatigue but not quality of life. Fasting, calorie-restricted, and anti-inflammatory diets did not significantly affect fatigue or quality of life, and ketogenic diets fell somewhere in between. GM appeared to be a pivotal element influenced not only by the type of diet but also by therapies. In fact, vitamin D supplementation, commonly prescribed for MS [121], may also influence gut bacterial populations by increasing the frequency of certain genera [122]. In vitro studies have shown that some MS drugs (e.g., fingolimod and teriflunomide) also have anti-inflammatory and antioxidant effects and can shape GM and inhibit the growth of neurotoxin-secreting gut bacteria. [123] Regular exercise has been shown to have positive effects on sleep, depression, paresthesia, fatigue, and cognitive performance [124,125]. However, the impact of physical activity on GM composition and microbial metabolites in the gastrointestinal tract in MS patients remains to be studied [126].
Other factors, such as socioeconomic status, quality of life, and personal motivation, may contribute to the uncertainty surrounding diet-related results. Adherence to an MS-specific diet is associated with higher socioeconomic status, better quality of life, and higher nutritional quality [127]. Enhancing personal motivation and ensuring positive support from study staff and family members are opportunities for future dietary intervention studies in MS, as they can improve adherence and reduce attrition [118,128]. Additionally, strategies should be developed to tailor study diets to the preferences of both individuals with MS and their household members to reduce feelings of burden and improve diet observance [118,128].
Progressive MS disabilities can impact grocery shopping, cooking, and eating, leading to weight loss, isolation, and dysphagia in advanced stages [129,130]. Proper nutritional support and guidance are necessary to ensure a correct diet at any age, starting from the early stages of the disease [120,131,132,133]. On the other hand, an unbalanced diet coupled with reduced physical activity can result in overweight/obesity, which triggers EAE and MS onset and progression [17,54,55,134,135,136,137,138]. Overweight and obesity are also associated with cardiovascular disease, which contributes to more rapid disability progression in MS [139,140,141].
Considering the role of GM in MS onset and progression, probiotics are of great interest [142]. Probiotic supplementation can modify GM composition and intestinal barrier function, potentially modulating GBA pathways, immune cells, and inflammatory cytokines. Studies in MS patients and animal models have shown promising but inconclusive results, including slower disability progression, reduced depressive symptoms, and improvements in general health [143,144]. Further research is needed to explore different strains and their effects on GM composition, as they may depend on ongoing diets and therapies [145,146,147,148].

4.2. Dietary Supplementations

Apart from probiotics, dietary supplementation of several compounds proposed to increase anti-inflammatory and antioxidant activities have been reviewed by a recent Cochrane metanalysis including 30 randomized controlled trials (RCT) or controlled clinical trials (CCT) among participants with MS on MS-related outcomes, i.e., relapses, disability progression, and magnetic resonance imaging (MRI) measures [149]. After reviewing dietary programs including supplementation to increase PUFAs [comparing PUFAs vs. MUFA or PUFA Omega 6 vs. Omega 3] and other dietary supplements (e.g., antioxidants, acetyl L-carnitine, biotin, creatine, riboflavin), the metanalysis concluded that, at present, there is insufficient evidence to determine whether supplementation with antioxidants or other dietary interventions has any real impact, whether beneficial or harmful, on MS-related outcomes [149,150]. These data confirmed others’ conclusions that the body of present evidence is primarily focused around the isolation of individual nutrients, many of which demonstrate no clear effect on major outcomes of MS progression [151]. Of note, although some uncertainties can depend on the dosages used and/or the duration of the treatments (e.g., PUFA), supplementations with other compounds (e.g., vitamin D) may depend on patients’ pre-existing nutritional adequacy as opposed to a need for high-dose supplementation [151]. Recent data confirm that although there are no statistically significant correlations between clinical outcomes and vitamin D serum levels or supplementations, patients receiving vitamin D had fewer new T2-weighted lesions, especially when optimal or higher levels of vitamin D (>30 ng/mL) were maintained throughout the entire 4-month observation period [152]. All in all, stronger studies focused on food and nutritional supplementation are required to strengthen the evidence.

5. Diet and Nutrition Related Issues in Pediatric-Onset MS

MS starting in childhood is estimated to account for between 2–5% and 5–10% of the MS population worldwide, with an estimated global incidence ranging from 0.05 to 2.85 per 100,000 children a year and a prevalence of 0.7 to 26.9 per 100,000 children. The median age of children with POMS is 12 years (range: 1.6–17). It is not clear whether the recently reported increased incidence may be real or more likely dependent on better diagnostic methods [2,153,154,155,156]. As outlined before, at any rate, data in POMS remain scarce mainly due to small sample size [1,157]. Despite POMS’s active inflammatory course, with frequent relapses/partial recovery episodes [158,159,160], presently available DMT may attain a discrete inflammation control [161,162], with a beneficial reduction of annualized relapse rates which may interfere with age specific cognitive impairment [160]. This is important because, although children and adolescents have a lower risk of disability within the first 10 years of diagnosis than those with adult-onset MS, in fact, the disease may negatively affect their school and emotional spheres [163]. Moreover, young patients reach disability milestones earlier than adults, even though they tend to take a longer time to advance to the secondary progressive phase [34,164,165]. Consequently, healthcare resource utilization and costs are high in this population compared with healthy children and adolescents. Quite common cognitive impairment requiring specific management, decrease in QOL, and an increase in economic burden in POMS have been shown to have profound impacts not only on patients but also on their families [154,155]. Still, an early onset of MS seems to be somewhat protective regarding QOL [165], maybe due to better resilience resources in youth [154,155,166]. Efforts towards a better understanding of possible specific mechanisms that trigger POMS and improving inherent therapies and prevention appear, therefore, to be necessary in young and pre-pubertal children [162,167,168].
Childhood obesity has been identified as a potential risk factor for increased morbidity not only from its hepatic-cardiac-metabolic comorbidities [169,170] but also from MS and clinically isolated syndrome (CIS) in adolescents, particularly in girls [134]. The underlying mechanism may involve vitamin D deficiency, as obesity is associated with lower vitamin D levels [136,171]. The association between excess weight and MS risk highlights the importance of addressing the childhood obesity epidemic and its potential impact on pediatric MS [171]. Of note, sedentary indoor lifestyles and reduced sunlight exposure, exacerbated by the COVID-19 pandemic, contribute to decreased vitamin D synthesis and increased hypovitaminosis D in children [172].
Dietary factors have also been studied in relation to MS outcomes in children. A study analyzing dietary intake in children with early-onset pediatric MS found that unbalanced diets with increased fats, especially saturated fat content, were associated with a higher risk of unfavorable disease progression [173]. However, sugar intake did not appear to be associated with a higher relapse risk [173]. Additionally, a healthy diet in childhood, characterized by the consumption of fruit, yogurt and legumes, was associated with a lower probability of developing MS in adulthood [174]. These findings suggest that specific dietary strategies may aid children with POMS in slowing disease progression and improving their quality of life [163].
Minerals, such as iron, may also play a role in POMS progression [175]. Iron deposition in gray and white matter has been observed in MS patients, and iron chelation therapy has been proposed as a potential therapeutic approach [176]. Conversely, iron deficiency may contribute to disease progression, as iron is involved in myelin synthesis and normal cellular metabolic pathways [177,178]. Further studies are needed to clarify the role of iron in POMS.
The gut microbiota (GM) has also been implicated in POMS. Differences in microbial composition and metabolic pathways have been observed in children with POMS compared to healthy controls [179,180]. GM composition may predict the likelihood of recurrence in pediatric MS [181,182].
Due to its effects on levels of disease activity, depression and fatigue, and lower moderate to vigorous physical activity, prevention and/or treatment of obesity/being overweight has been deemed a critical aspect of POMS as well [183]. A case-control study comparing the BMI data of 60 French youths with POMS showed that overweight and obesity are more frequently observed at diagnosis, particularly in boys with POMS compared with non-neurologic controls and healthy French children. Moreover, higher BMI is related to initial inflammation in the CSF in prepubertal patients with POMS suggesting an interaction between excess body fat, sexual hormones, and POMS occurrence [135]. These data are particularly worrying if one considers the findings that a large proportion of adolescents with POMS also have a non-self-perceived elevated BMI [184]. To improve their disease progression, they should therefore receive more accurate counseling to improve their diet [184] and physical activity as well [185].

6. Discussion

The last few years have been characterized by a mounting number of articles providing experimental and clinical evidence of the role of some nutrients and diets in MS. Most studies have regarded adult MS, focusing prevalently on risk factors rather than on disease progression, and there is still limited available evidence for many foods and nutrients [186]. Although more research is needed to fully understand their relationship with MS, vitamin D and PUFA appear to be the most relevant single molecules researched. Studies suggest that people with lower vitamin D levels have a higher risk of developing MS. In addition, research has shown that vitamin D supplementation can help reduce the severity and frequency of MS symptoms. Similarly, a diet high in PUFAs can help reduce the risk of developing MS and improve symptoms in people who already have the disease. Other relevant protective dietary factors could start during the early years, such as a prolonged duration of breastfeeding [187,188,189]. Regarding diets, many suggestions have been reported over time. Granted that no universal best diet exists, as it can vary based on patients’ individual needs and preferences, and that healthy and balanced diets are necessary to ensure adequate intake of all essential nutrients, some diets that have shown some beneficial effects typically share the conditions of having high intakes of fruits, vegetables, whole grains, and lean proteins while limiting processed foods, sugar, and saturated fat. All in all, these components have antioxidant and anti-inflammatory properties and tend to shape a healthy GM capable of respecting the gut and brain barriers. The effects of a Mediterranean diet on MS disability appear appealing but remain still uncertain [119,190]. Several other nutrition-related strategies have been proposed as an additional measure to DMTs. Among these, probiotics in the future will need further, better-conducted studies to confirm whether they can represent a viable chance as a personalized microbiota-based adjuvant therapy. Nondigestible dietary fibers (prebiotics) offer another related possibility to reshape altered GM in MS, which appears to be a practicable avenue warranting to be further explored. In fact, they contribute to gut eubiosis allowing the formation of microbial metabolites such as SCFAs in the colonic lumen with key immunological regulatory mechanisms in MS [29,191].
So far, nutritional and dietary data available on POMS are limited. However, they suggest that, as in adults, POMS’ disease management might benefit from an add-on nutritional intervention. In particular, it appears that a diet rich in vegetable intake and low in saturated fat may reduce the risk of relapse [173]. Studies on other interventions for this group are necessary, because simple translation of evidence targeting diet or nutrition available in adults is not always feasible, as it may be the case of salt intake [192,193,194,195]. Besides dietary factors, physical activity (PA) and sleep are the other two important health behaviors in both adults and youths with MS, as they represent a modifiable factor. In a scoping review of the available literature [195], the authors concluded that the PA self-reported by children with POMS was (a) generally lower than that of healthy controls and those with other types of demyelinating diseases, and (b) may be associated with markers of disease burden and symptomatology. Intriguingly, children with POMS performed better than matched controls in managing sleep hygiene, and higher levels of physical activity were associated with fewer sleep/rest fatigue symptoms [195]. It is noteworthy that, quite similar conclusions were also reached by other independent authors [183].
Overall, our review must take account of its limitations due to the fact that only a few of the considered studies were controlled and the enrolled patients who volunteered for randomization were obviously not blind. Both in adults and in pediatric age, most data derive from enquiry-based dietary/nutritional studies which have, as it may be expected, several drawbacks. These include the fact that food frequency questionnaires may be subject to recall-bias-driven report errors, lack of relevant food items and nutrients, and focus on a few days’ diet instead of real long-term dietetic habits. Moreover, their retrospective nature only allows them to shed light on an association and not on a causation [80,112,190]. On top of these issues, confounding conclusions may derive from interferences deriving from the real level of adherence to the diets [127,128,129,130,196], level of sleep [124,125], and the quite under-rated effects of associated PA [124,125,126], vitamin D [122] and some DMT [123] on GM.
Finally, new, yet unexplored therapeutical paths are mandatory to further complement the presently available therapies. What is around the corner? Next generation (NG) engineered probiotics, obtained by modifying original probiotics through gene editing modalities, have hitherto been used in inflammatory bowel disease, and in a number of bacterial infections, tumors, and metabolic diseases, mainly in EAE murine models and/or in vitro [197]. Promising preliminary results showing they are effective, with fewer side effects than traditional treatments or wild-type strains [198,199], suggest that they will probably be proposed soon for CNS diseases as well, including MS. Of note, it has been underlined that the design of NG probiotics should specifically be directed towards increasing the production of metabolites (e.g., SCFAs) and neurotransmitters (e.g., serotonin, GABA) which are known to affect the neurobiology of CNS inflammatory diseases [200]. Last but not least, fecal microbiota transplantation (FMT) represents a further interesting approach to modulate GM [201,202]. FMT studies in animal models and in humans with MS are still scarce and preliminary. Some data available from a cohort of RRMS patients show that FMT is safe and was well-tolerated and may have also improved their gut dysbiosis and elevated small intestinal permeability [203]. Moreover, single case reports and a case series (totaling six patients) reviewed by Matheson et al., in addition to confirming the safety of the treatment, also showed specific clinical improvements in MS-related neurological symptoms [204].

7. Conclusions

In conclusion, several dietary/nutritional factors play an important role in MS development and progression. Several gut-oriented nutritional interventions aiming to improve the dysregulation of the so-called gut-brain axis through a proper diet appear to intervene beneficially mainly against the inflammatory pathomechanisms associated with MS. Nutritional issues are therefore worthy of further research with high-quality, long-term, randomized studies in the global treatment of patients with MS [205]. The efficacy of any dietary intervention in MS, however, remains very difficult to prove due to spontaneous remissions (and relapses) with temporary clinical improvement occurring by chance alone. Pending more solid evidence on specific diets, experts suggest that individuals with MS should be taught to follow a “healthy” regime [128,190] and possibly enter into nutrition education programs, which, however, are largely lacking for patients with MS and most other neurological diseases at present [206]. Because of the high prevalence of overweight/obesity, and the evidence that obesity can worsen MS prognosis, education on weight management is still an unmet need, and non-guided patients are frequently no more likely to adopt a specific diet than normal weight patients [138]. Pediatric interventions may be even more hampered by the lack of self-perceived BMI elevation in MS youths [184,185]. Because there is, at present, no robust evidence, future research is also needed to identify appropriate study designs and intervention strategies targeting PA participation, with measurements of their outcomes on the primary and secondary symptoms of POMS [195]. New solid longitudinal and experimental designs are necessary not only to better elucidate the role of diet and other modifiable lifestyle factors in this population, but also to explore other modalities of support. These should include a closer monitoring of nutritional status of patients with moderate-advanced MS in order to prevent their tendency to be overweight secondary to the decrease in basal energy expenditure and loss of muscle mass. Additionally, the intervention of other health/psychology/social professionals should contribute to overcoming barriers and improving quality of life by avoiding MS patients’ and their relatives’ struggles with loneliness and rejection [132,133].

Author Contributions

Conceptualization, C.M., R.L., A.C. and P.V.; methodology, A.C., M.R.S., J.S.N. and G.M.; writing—original sections of the draft preparation, C.M., R.L., A.C., E.S., C.D.M., L.S. and F.F.O.; writing—review and editing, C.M., A.C. and R.L.; supervision, C.M, A.S., L.S. and P.V.; project administration, P.V., C.M. and A.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study did not require ethical approval. Please note that the Editorial Office might ask you for further information.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge Anna Pia Delli Bovi for her help in organizing the references of the first draft. We acknowledge Annalisa Morelli for the language revision.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Alroughani, R.; Boyko, A. Pediatric Multiple Sclerosis: A Review. BMC Neurol. 2018, 18, 27. [Google Scholar] [CrossRef] [PubMed]
  2. Yan, K.; Balijepalli, C.; Desai, K.; Gullapalli, L.; Druyts, E. Epidemiology of Pediatric Multiple Sclerosis: A Systematic Literature Review and Meta-Analysis. Mult. Scler. Relat. Disord. 2020, 44, 102260. [Google Scholar] [CrossRef] [PubMed]
  3. Umeton, R.; Bellucci, G.; Bigi, R.; Romano, S.; Buscarinu, M.C.; Reniè, R.; Rinaldi, V.; Pizzolato Umeton, R.; Morena, E.; Romano, C. Multiple Sclerosis Genetic and Non-Genetic Factors Interact through the Transient Transcriptome. Sci. Rep. 2022, 12, 7536. [Google Scholar] [CrossRef] [PubMed]
  4. Natrajan, M.S.; de la Fuente, A.G.; Crawford, A.H.; Linehan, E.; Nunez, V.; Johnson, K.R.; Wu, T.; Fitzgerald, D.C.; Ricote, M.; Bielekova, B. Retinoid X Receptor Activation Reverses Age-Related Deficiencies in Myelin Debris Phagocytosis and Remyelination. Brain 2015, 138, 3581–3597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Tafti, D.; Ehsan, M.; Xixis, K.L. Multiple Sclerosis. Available online: https://www.ncbi.nlm.nih.gov/books/NBK499849/ (accessed on 7 May 2023).
  6. Balcerac, A.; Louapre, C. Genetics and Familial Distribution of Multiple Sclerosis: A Review. Rev. Neurol. 2022, 178, 512–520. [Google Scholar] [CrossRef]
  7. Huang, J.; Kockum, I.; Stridh, P. Trends in the Environmental Risks Associated with Earlier Onset in Multiple Sclerosis. Mult. Scler. Relat. Disord. 2022, 68, 104250. [Google Scholar] [CrossRef]
  8. Walton, C.; King, R.; Rechtman, L.; Kaye, W.; Leray, E.; Marrie, R.A.; Robertson, N.; La Rocca, N.; Uitdehaag, B.; van Der Mei, I. Rising Prevalence of Multiple Sclerosis Worldwide: Insights from the Atlas of MS. Mult. Scler. J. 2020, 26, 1816–1821. [Google Scholar] [CrossRef]
  9. Beckett, J.M.; Bird, M.-L.; Pittaway, J.K.; Ahuja, K.D. Diet and Multiple Sclerosis: Scoping Review of Web-Based Recommendations. Interact. J. Med. Res. 2019, 8, e10050. [Google Scholar] [CrossRef]
  10. Disease Modifying Therapies. Available online: https://www.mssociety.org.uk/about-ms/treatments-and-therapies/disease-modifying-therapies (accessed on 7 May 2023).
  11. Stoiloudis, P.; Kesidou, E.; Bakirtzis, C.; Sintila, S.-A.; Konstantinidou, N.; Boziki, M.; Grigoriadis, N. The Role of Diet and Interventions on Multiple Sclerosis: A Review. Nutrients 2022, 14, 1150. [Google Scholar] [CrossRef]
  12. Riccio, P.; Rossano, R. Diet, Gut Microbiota, and Vitamins D + A in Multiple Sclerosis. Neurotherapeutics 2018, 15, 75–91. [Google Scholar] [CrossRef] [Green Version]
  13. Parodi, B.; Kerlero de Rosbo, N. The Gut-Brain Axis in Multiple Sclerosis. Is Its Dysfunction a Pathological Trigger or a Consequence of the Disease? Front. Immunol. 2021, 12, 718220. [Google Scholar] [CrossRef] [PubMed]
  14. Bierhansl, L.; Hartung, H.-P.; Aktas, O.; Ruck, T.; Roden, M.; Meuth, S.G. Thinking Outside the Box: Non-Canonical Targets in Multiple Sclerosis. Nat. Rev. Drug. Discov. 2022, 21, 578–600. [Google Scholar] [CrossRef] [PubMed]
  15. Timmermans, S.; Bogie, J.F.; Vanmierlo, T.; Lütjohann, D.; Stinissen, P.; Hellings, N.; Hendriks, J.J. High Fat Diet Exacerbates Neuroinflammation in an Animal Model of Multiple Sclerosis by Activation of the Renin Angiotensin System. J. Neuroimmune Pharmacol. 2014, 9, 209–217. [Google Scholar] [CrossRef]
  16. Yoon, H.; Kleven, A.; Paulsen, A.; Kleppe, L.; Wu, J.; Ying, Z.; Gomez-Pinilla, F.; Scarisbrick, I.A. Interplay between Exercise and Dietary Fat Modulates Myelinogenesis in the Central Nervous System. Biochim. Biophys. Acta BBA-Mol. Basis Dis. 2016, 1862, 545–555. [Google Scholar] [CrossRef] [PubMed]
  17. Guerrero-García, J.d.J.; Carrera-Quintanar, L.; López-Roa, R.I.; Márquez-Aguirre, A.L.; Rojas-Mayorquín, A.E.; Ortuño-Sahagún, D. Multiple Sclerosis and Obesity: Possible Roles of Adipokines. Mediators Inflamm. 2016, 2016, 4036232. [Google Scholar] [CrossRef] [Green Version]
  18. Nyirenda, M.H.; Fadda, G.; Healy, L.M.; Mexhitaj, I.; Poliquin-Lasnier, L.; Hanwell, H.; Saveriano, A.W.; Rozenberg, A.; Li, R.; Moore, C.S. Pro-Inflammatory Adiponectin in Pediatric-Onset Multiple Sclerosis. Mult. Scler. J. 2021, 27, 1948–1959. [Google Scholar] [CrossRef]
  19. Paolella, G.; Mandato, C.; Pierri, L.; Poeta, M.; Di Stasi, M.; Vajro, P. Gut-Liver Axis and Probiotics: Their Role in Non-Alcoholic Fatty Liver Disease. World J. Gastroenterol. WJG 2014, 20, 15518. [Google Scholar] [CrossRef] [PubMed]
  20. Mandato, C.; Bovi, A.P.D.; Vajro, P. The Gut-Liver Axis as a Target of Liver Disease Management. Hepatobiliary Surg. Nutr. 2021, 10, 100. [Google Scholar] [CrossRef] [PubMed]
  21. Spadoni, I.; Zagato, E.; Bertocchi, A.; Paolinelli, R.; Hot, E.; Di Sabatino, A.; Caprioli, F.; Bottiglieri, L.; Oldani, A.; Viale, G. A Gut-Vascular Barrier Controls the Systemic Dissemination of Bacteria. Science 2015, 350, 830–834. [Google Scholar] [CrossRef]
  22. Di Tommaso, N.; Santopaolo, F.; Gasbarrini, A.; Ponziani, F.R. The Gut–Vascular Barrier as a New Protagonist in Intestinal and Extraintestinal Diseases. Int. J. Mol. Sci. 2023, 24, 1470. [Google Scholar] [CrossRef]
  23. Camara-Lemarroy, C.R.; Metz, L.M.; Yong, V.W. Focus on the Gut-Brain Axis: Multiple Sclerosis, the Intestinal Barrier and the Microbiome. World J. Gastroenterol. 2018, 24, 4217. [Google Scholar] [CrossRef]
  24. Boziki, M.K.; Kesidou, E.; Theotokis, P.; Mentis, A.-F.A.; Karafoulidou, E.; Melnikov, M.; Sviridova, A.; Rogovski, V.; Boyko, A.; Grigoriadis, N. Microbiome in Multiple Sclerosis: Where Are We, What We Know and Do Not Know. Brain Sci. 2020, 10, 234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Panera, N.; Mandato, C.; Crudele, A.; Bertrando, S.; Vajro, P.; Alisi, A. Genetics, Epigenetics and Transgenerational Transmission of Obesity in Children. Front. Endocrinol. 2022, 13, 1006008. [Google Scholar] [CrossRef] [PubMed]
  26. Madison, A.; Kiecolt-Glaser, J.K. Stress, Depression, Diet, and the Gut Microbiota: Human–Bacteria Interactions at the Core of Psychoneuroimmunology and Nutrition. Curr. Opin. Behav. Sci. 2019, 28, 105–110. [Google Scholar] [CrossRef] [PubMed]
  27. Altowaijri, G.; Fryman, A.; Yadav, V. Dietary Interventions and Multiple Sclerosis. Curr. Neurol. Neurosci. Rep. 2017, 17, 28. [Google Scholar] [CrossRef]
  28. Freedman, S.N.; Shahi, S.K.; Mangalam, A.K. The “Gut Feeling”: Breaking down the Role of Gut Microbiome in Multiple Sclerosis. Neurotherapeutics 2018, 15, 109–125. [Google Scholar] [CrossRef] [Green Version]
  29. Ullah, H.; Tovchiga, O.; Daglia, M.; Khan, H. Modulating Gut Microbiota: An Emerging Approach in the Prevention and Treatment of Multiple Sclerosis. Curr. Neuropharmacol. 2021, 19, 1966. [Google Scholar] [CrossRef]
  30. Sorboni, S.G.; Moghaddam, H.S.; Jafarzadeh-Esfehani, R.; Soleimanpour, S. A Comprehensive Review on the Role of the Gut Microbiome in Human Neurological Disorders. Clin. Microbiol. Rev. 2022, 35, e00338-20. [Google Scholar] [CrossRef]
  31. Wang, X.; Liang, Z.; Wang, S.; Ma, D.; Zhu, M.; Feng, J. Role of Gut Microbiota in Multiple Sclerosis and Potential Therapeutic Implications. Curr. Neuropharmacol. 2022, 20, 1413–1426. [Google Scholar] [CrossRef]
  32. Preiningerova, J.L.; Jiraskova Zakostelska, Z.; Srinivasan, A.; Ticha, V.; Kovarova, I.; Kleinova, P.; Tlaskalova-Hogenova, H.; Kubala Havrdova, E. Multiple Sclerosis and Microbiome. Biomolecules 2022, 12, 433. [Google Scholar] [CrossRef]
  33. Engelenburg, H.J.; Lucassen, P.J.; Sarafian, J.T.; Parker, W.; Laman, J.D. Multiple Sclerosis and the Microbiota: Progress in Understanding the Contribution of the Gut Microbiome to Disease. Evol. Med. Public Health 2022, 10, 277–294. [Google Scholar] [CrossRef] [PubMed]
  34. Yu, H.; Bai, S.; Hao, Y.; Guan, Y. Fatty Acids Role in Multiple Sclerosis as “Metabokines”. J. Neuroinflammation 2022, 19, 157. [Google Scholar] [CrossRef] [PubMed]
  35. Nouri, M.; Bredberg, A.; Weström, B.; Lavasani, S. Intestinal Barrier Dysfunction Develops at the Onset of Experimental Autoimmune Encephalomyelitis, and Can Be Induced by Adoptive Transfer of Auto-Reactive T Cells. PLoS ONE 2014, 9, e106335. [Google Scholar] [CrossRef] [PubMed]
  36. Dargahi, N.; Matsoukas, J.; Apostolopoulos, V. Streptococcus Thermophilus ST285 Alters Pro-Inflammatory to Anti-Inflammatory Cytokine Secretion against Multiple Sclerosis Peptide in Mice. Brain Sci. 2020, 10, 126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Cani, P.D.; de Vos, W.M. Next-Generation Beneficial Microbes: The Case of Akkermansia Muciniphila. Front. Microbiol. 2017, 8, 1765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Xu, R.; Zhang, Y.; Chen, S.; Zeng, Y.; Fu, X.; Chen, T.; Luo, S.; Zhang, X. The Role of the Probiotic Akkermansia Muciniphila in Brain Functions: Insights Underpinning Therapeutic Potential. Crit. Rev. Microbiol. 2023, 49, 151–176. [Google Scholar] [CrossRef]
  39. Shahi, S.K.; Freedman, S.N.; Mangalam, A.K. Gut Microbiome in Multiple Sclerosis: The Players Involved and the Roles They Play. Gut Microbes 2017, 8, 607–615. [Google Scholar] [CrossRef] [Green Version]
  40. Farrokhi, V.; Nemati, R.; Nichols, F.C.; Yao, X.; Anstadt, E.; Fujiwara, M.; Grady, J.; Wakefield, D.; Castro, W.; Donaldson, J. Bacterial Lipodipeptide, Lipid 654, Is a Microbiome-associated Biomarker for Multiple Sclerosis. Clin. Transl. Immunol. 2013, 2, e8. [Google Scholar] [CrossRef] [PubMed]
  41. Ling, Z.; Cheng, Y.; Yan, X.; Shao, L.; Liu, X.; Zhou, D.; Zhang, L.; Yu, K.; Zhao, L. Alterations of the Fecal Microbiota in Chinese Patients with Multiple Sclerosis. Front. Immunol. 2020, 11, 590783. [Google Scholar] [CrossRef]
  42. Dias de Sousa, M.A.; Desidério, C.S.; da Silva Catarino, J.; Trevisan, R.O.; Alves da Silva, D.A.; Rocha, V.F.R.; Bovi, W.G.; Timoteo, R.P.; Bonatti, R.C.F.; da Silva, A.E. Role of Cytokines, Chemokines and IFN-Γ+ IL-17+ Double-Positive CD4+ T Cells in Patients with Multiple Sclerosis. Biomedicines 2022, 10, 2062. [Google Scholar] [CrossRef]
  43. Duarte-Silva, E.; Meuth, S.G.; Peixoto, C.A. Microbial Metabolites in Multiple Sclerosis: Implications for Pathogenesis and Treatment. Front. Neurosci. 2022, 16, 885031. [Google Scholar] [CrossRef] [PubMed]
  44. Levi, I.; Gurevich, M.; Perlman, G.; Magalashvili, D.; Menascu, S.; Bar, N.; Godneva, A.; Zahavi, L.; Chermon, D.; Kosower, N. Potential Role of Indolelactate and Butyrate in Multiple Sclerosis Revealed by Integrated Microbiome-Metabolome Analysis. Cell Rep. Med. 2021, 2, 100246. [Google Scholar] [CrossRef] [PubMed]
  45. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [Green Version]
  46. Takewaki, D.; Suda, W.; Sato, W.; Takayasu, L.; Kumar, N.; Kimura, K.; Kaga, N.; Mizuno, T.; Miyake, S.; Hattori, M. Alterations of the Gut Ecological and Functional Microenvironment in Different Stages of Multiple Sclerosis. Proc. Natl. Acad. Sci. USA 2020, 117, 22402–22412. [Google Scholar] [CrossRef] [PubMed]
  47. Fyfe, I. Microbiome Varies between Multiple Sclerosis Subtypes. Nat. Rev. Neurol. 2020, 16, 243. [Google Scholar] [CrossRef] [Green Version]
  48. Reynders, T.; Devolder, L.; Valles-Colomer, M.; Van Remoortel, A.; Joossens, M.; De Keyser, J.; Nagels, G.; D’hooghe, M.; Raes, J. Gut Microbiome Variation Is Associated to Multiple Sclerosis Phenotypic Subtypes. Ann. Clin. Transl. Neurol. 2020, 7, 406–419. [Google Scholar] [CrossRef] [Green Version]
  49. Cekanaviciute, E.; Yoo, B.B.; Runia, T.F.; Debelius, J.W.; Singh, S.; Nelson, C.A.; Kanner, R.; Bencosme, Y.; Lee, Y.K.; Hauser, S.L. Gut Bacteria from Multiple Sclerosis Patients Modulate Human T Cells and Exacerbate Symptoms in Mouse Models. Proc. Natl. Acad. Sci. USA 2017, 114, 10713–10718. [Google Scholar] [CrossRef] [Green Version]
  50. Berer, K.; Gerdes, L.A.; Cekanaviciute, E.; Jia, X.; Xiao, L.; Xia, Z.; Liu, C.; Klotz, L.; Stauffer, U.; Baranzini, S.E. Gut Microbiota from Multiple Sclerosis Patients Enables Spontaneous Autoimmune Encephalomyelitis in Mice. Proc. Natl. Acad. Sci. USA 2017, 114, 10719–10724. [Google Scholar] [CrossRef] [Green Version]
  51. Luu, M.; Pautz, S.; Kohl, V.; Singh, R.; Romero, R.; Lucas, S.; Hofmann, J.; Raifer, H.; Vachharajani, N.; Carrascosa, L.C. The Short-Chain Fatty Acid Pentanoate Suppresses Autoimmunity by Modulating the Metabolic-Epigenetic Crosstalk in Lymphocytes. Nat. Commun. 2019, 10, 760. [Google Scholar] [CrossRef] [Green Version]
  52. Cantoni, C.; Lin, Q.; Dorsett, Y.; Ghezzi, L.; Liu, Z.; Pan, Y.; Chen, K.; Han, Y.; Li, Z.; Xiao, H. Alterations of Host-Gut Microbiome Interactions in Multiple Sclerosis. EBioMedicine 2022, 76, 103798. [Google Scholar] [CrossRef]
  53. Kadry, H.; Noorani, B.; Cucullo, L. A Blood–Brain Barrier Overview on Structure, Function, Impairment, and Biomarkers of Integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef]
  54. Davanzo, G.G.; Castro, G.; de Brito Monteiro, L.; Castelucci, B.G.; Jaccomo, V.H.; da Silva, F.C.; Marques, A.M.; Francelin, C.; de Campos, B.B.; de Aguiar, C.F. Obesity Increases Blood-Brain Barrier Permeability and Aggravates the Mouse Model of Multiple Sclerosis. Mult. Scler. Relat. Disord. 2023, 72, 104605. [Google Scholar] [CrossRef] [PubMed]
  55. Ji, Z.; Wu, S.; Xu, Y.; Qi, J.; Su, X.; Shen, L. Obesity Promotes EAE through IL-6 and CCL-2-Mediated T Cells Infiltration. Front. Immunol. 2019, 10, 1881. [Google Scholar] [CrossRef] [Green Version]
  56. Babaloo, Z.; Aliparasti, M.R.; Babaiea, F.; Almasi, S.; Baradaran, B.; Farhoudi, M. The Role of Th17 Cells in Patients with Relapsing-Remitting Multiple Sclerosis: Interleukin-17A and Interleukin-17F Serum Levels. Immunol. Lett. 2015, 164, 76–80. [Google Scholar] [CrossRef]
  57. Awasthi, A.; Kuchroo, V.K. Th17 Cells: From Precursors to Players in Inflammation and Infection. Int. Immunol. 2009, 21, 489–498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Li, X.; You, X.; Wang, C.; Li, X.; Sheng, Y.; Zhuang, P.; Zhang, Y. Bidirectional Brain-gut-microbiota Axis in Increased Intestinal Permeability Induced by Central Nervous System Injury. CNS Neurosci. Ther. 2020, 26, 783–790. [Google Scholar] [CrossRef]
  59. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The Gut-Brain Axis: Interactions between Enteric Microbiota, Central and Enteric Nervous Systems. Ann. Gastroenterol. Q. Publ. Hell. Soc. Gastroenterol. 2015, 28, 203. [Google Scholar]
  60. Abdollahpour, I.; Jakimovski, D.; Shivappa, N.; Hébert, J.R.; Vahid, F.; Nedjat, S.; Mansournia, M.A.; Weinstock-Guttman, B. Dietary Inflammatory Index and Risk of Multiple Sclerosis: Findings from a Large Population-Based Incident Case–Control Study. Clin. Nutr. 2020, 39, 3402–3407. [Google Scholar] [CrossRef]
  61. Alhaj, O.A.; Trabelsi, K.; Younes, A.M.; Shivappa, N.; Bragazzi, N.L.; Hebert, J.R.; Jahrami, H.A. Diet-Related Inflammation Increases the Odds of Multiple Sclerosis: Results from a Large Population-Based Prevalent Case-Control Study in Jordan. Front. Nutr. 2023, 10, 554. [Google Scholar] [CrossRef]
  62. Hoare, S.; Lithander, F.; Van Der Mei, I.; Ponsonby, A.-L.; Lucas, R.; Group, A.I. Higher Intake of Omega-3 Polyunsaturated Fatty Acids Is Associated with a Decreased Risk of a First Clinical Diagnosis of Central Nervous System Demyelination: Results from the Ausimmune Study. Mult. Scler. J. 2016, 22, 884–892. [Google Scholar] [CrossRef]
  63. Matveeva, O.; Bogie, J.F.; Hendriks, J.J.; Linker, R.A.; Haghikia, A.; Kleinewietfeld, M. Western Lifestyle and Immunopathology of Multiple Sclerosis. Ann. N. Y. Acad. Sci. 2018, 1417, 71–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Leo, E.E.M.; Campos, M.R.S. Effect of Ultra-Processed Diet on Gut Microbiota and Thus Its Role in Neurodegenerative Diseases. Nutrition 2020, 71, 110609. [Google Scholar] [CrossRef]
  65. Mannino, A.; Daly, A.; Dunlop, E.; Probst, Y.; Ponsonby, A.-L.; van der Mei, I.A.F.; Chapman, C.; Coulthard, A.; Dear, K.; Dwyer, T.; et al. Higher Consumption of Ultra-Processed Foods and Increased Likelihood of Central Nervous System Demyelination in a Case-Control Study of Australian Adults. Eur. J. Clin. Nutr. 2023, 77, 611–614. [Google Scholar] [CrossRef] [PubMed]
  66. Martín-Hersog, F.A.; Muñoz-Jurado, A.; Escribano, B.M.; Luque, E.; Galván, A.; LaTorre, M.; Giraldo, A.I.; Caballero-Villarraso, J.; Agüera, E.; Santamaría, A. Sodium Chloride-Induced Changes in Oxidative Stress, Inflammation, and Dysbiosis in Experimental Multiple Sclerosis. Nutr. Neurosci. 2022, 1–13. [Google Scholar] [CrossRef] [PubMed]
  67. Kleinewietfeld, M.; Manzel, A.; Titze, J.; Kvakan, H.; Yosef, N.; Linker, R.A.; Muller, D.N.; Hafler, D.A. Sodium Chloride Drives Autoimmune Disease by the Induction of Pathogenic TH17 Cells. Nature 2013, 496, 518–522. [Google Scholar] [CrossRef] [Green Version]
  68. Farez, M.F.; Fiol, M.P.; Gaitán, M.I.; Quintana, F.J.; Correale, J. Sodium Intake Is Associated with Increased Disease Activity in Multiple Sclerosis. J. Neurol. Neurosurg. Psychiatry 2015, 86, 26–31. [Google Scholar] [CrossRef] [Green Version]
  69. Riccio, P.; Rossano, R. Undigested Food and Gut Microbiota May Cooperate in the Pathogenesis of Neuroinflammatory Diseases: A Matter of Barriers and a Proposal on the Origin of Organ Specificity. Nutrients 2019, 11, 2714. [Google Scholar] [CrossRef] [Green Version]
  70. Black, L.J.; Zhao, Y.; Peng, Y.C.; Sherriff, J.L.; Lucas, R.M.; Van Der Mei, I.; Pereira, G.; The Ausimmune Investigator Group. Higher Fish Consumption and Lower Risk of Central Nervous System Demyelination. Eur. J. Clin. Nutr. 2020, 74, 818–824. [Google Scholar] [CrossRef]
  71. Bayat, P.; Farshchi, M.; Yousefian, M.; Mahmoudi, M.; Yazdian-Robati, R. Flavonoids, the Compounds with Anti-Inflammatory and Immunomodulatory Properties, as Promising Tools in Multiple Sclerosis (MS) Therapy: A Systematic Review of Preclinical Evidence. Int. Immunopharmacol. 2021, 95, 107562. [Google Scholar] [CrossRef]
  72. Hashimoto, M.; Yamamoto, S.; Iwasa, K.; Yamashina, K.; Ishikawa, M.; Maruyama, K.; Bosetti, F.; Yoshikawa, K. The Flavonoid Baicalein Attenuates Cuprizone-Induced Demyelination via Suppression of Neuroinflammation. Brain Res. Bull. 2017, 135, 47–52. [Google Scholar] [CrossRef]
  73. Evans, E.; Levasseur, V.; Cross, A.H.; Piccio, L. An Overview of the Current State of Evidence for the Role of Specific Diets in Multiple Sclerosis. Mult. Scler. Relat. Disord. 2019, 36, 101393. [Google Scholar] [CrossRef]
  74. Marck, C.H.; Probst, Y.; Chen, J.; Taylor, B.; van der Mei, I. Dietary Patterns and Associations with Health Outcomes in Australian People with Multiple Sclerosis. Eur. J. Clin. Nutr. 2021, 75, 1506–1514. [Google Scholar] [CrossRef]
  75. Yadav, V.; Marracci, G.; Kim, E.; Spain, R.; Cameron, M.; Overs, S.; Riddehough, A.; Li, D.K.; McDougall, J.; Lovera, J. Low-Fat, Plant-Based Diet in Multiple Sclerosis: A Randomized Controlled Trial. Mult. Scler. Relat. Disord. 2016, 9, 80–90. [Google Scholar] [CrossRef] [Green Version]
  76. Ashtari, F.; Jamshidi, F.; Shoormasti, R.S.; Pourpak, Z.; Akbari, M. Cow’s Milk Allergy in Multiple Sclerosis Patients. J. Res. Med. Sci. Off. J. Isfahan Univ. Med. Sci. 2013, 18, S62. [Google Scholar]
  77. Stefferl, A.; Schubart, A.; Storch, M.; Amini, A.; Mather, I.; Lassmann, H.; Linington, C. Butyrophilin, a Milk Protein, Modulates the Encephalitogenic T Cell Response to Myelin Oligodendrocyte Glycoprotein in Experimental Autoimmune Encephalomyelitis. J. Immunol. 2000, 165, 2859–2865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Guggenmos, J.; Schubart, A.S.; Ogg, S.; Andersson, M.; Olsson, T.; Mather, I.H.; Linington, C. Antibody Cross-Reactivity between Myelin Oligodendrocyte Glycoprotein and the Milk Protein Butyrophilin in Multiple Sclerosis. J. Immunol. 2004, 172, 661–668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Hadgkiss, E.J.; Jelinek, G.A.; Weiland, T.J.; Pereira, N.G.; Marck, C.H.; van der Meer, D.M. The Association of Diet with Quality of Life, Disability, and Relapse Rate in an International Sample of People with Multiple Sclerosis. Nutr. Neurosci. 2015, 18, 125–136. [Google Scholar] [CrossRef] [Green Version]
  80. Fitzgerald, K.C.; Tyry, T.; Salter, A.; Cofield, S.S.; Cutter, G.; Fox, R.; Marrie, R.A. Diet Quality Is Associated with Disability and Symptom Severity in Multiple Sclerosis. Neurology 2018, 90, e1–e11. [Google Scholar] [CrossRef]
  81. Passali, M.; Josefsen, K.; Frederiksen, J.L.; Antvorskov, J.C. Current Evidence on the Efficacy of Gluten-Free Diets in Multiple Sclerosis, Psoriasis, Type 1 Diabetes and Autoimmune Thyroid Diseases. Nutrients 2020, 12, 2316. [Google Scholar] [CrossRef]
  82. Thomsen, H.L.; Jessen, E.B.; Passali, M.; Frederiksen, J.L. The Role of Gluten in Multiple Sclerosis: A Systematic Review. Mult. Scler. Relat. Disord. 2019, 27, 156–163. [Google Scholar] [CrossRef] [PubMed]
  83. Rodrigo, L.; Hernández-Lahoz, C.; Fuentes, D.; Mauri, G.; Alvarez, N.; Vega, J.; González, S. Randomised Clinical Trial Comparing the Efficacy of a Gluten-Free Diet versus a Regular Diet in a Series of Relapsing-Remitting Multiple Sclerosis Patients. Int. J. Neurol. Neurother. 2014, 1, 1–6. [Google Scholar] [CrossRef]
  84. Peng, H.; Wu, X.; Wen, Y.; Lin, J. Plasma Circulating Vitamin C Levels and Risk of Multiple Sclerosis: A Two-Sample Mendelian Randomization Analysis. Mult. Scler. Relat. Disord. 2021, 56, 103267. [Google Scholar] [CrossRef] [PubMed]
  85. Bowling, A. Vitamins, Minerals & Herbs in MS AN INTRODUCTION. Available online: https://www.nationalmssociety.org/Programs-and-Services/Resources/Vitamins,-Minerals,-and-Herbs-in-MS-An-Introductio (accessed on 6 May 2023).
  86. Zorzella-Pezavento, S.F.G.; Mimura, L.A.N.; Denadai, M.B.; De Souza, W.D.F.; de Campos Fraga-Silva, T.F.; Sartori, A. Is There a Window of Opportunity for the Therapeutic Use of Vitamin D in Multiple Sclerosis? Neural Regen. Res. 2022, 17, 1945. [Google Scholar] [CrossRef] [PubMed]
  87. Vandebergh, M.; Dubois, B.; Goris, A. Effects of Vitamin D and Body Mass Index on Disease Risk and Relapse Hazard in Multiple Sclerosis: A Mendelian Randomization Study. Neurol.-Neuroimmunol. Neuroinflammation 2022, 9, e1165. [Google Scholar] [CrossRef]
  88. Hebener, O.; Ackermann, H.; Kappel, U.; Kramer, J. Multiple Sclerosis and Additional Balanced Diet. Results of a Retrospective Study. Schweiz. Z. Für Ganzheitsmed./Swiss J. Integr. Med. 2005, 7, 104–108. [Google Scholar]
  89. Kousmine, C. Sauvez Votre Corps; Robert Laffont: Paris, France, 1987; ISBN 13: 9782221053843. [Google Scholar]
  90. Newman, P.E. Could Diet Be One of the Causal Factors of Alzheimer’s Disease? Med. Hypotheses 1992, 39, 123–126. [Google Scholar] [CrossRef] [PubMed]
  91. Farinotti, M.; Vacchi, L.; Simi, S.; Di Pietrantonj, C.; Brait, L.; Filippini, G. Dietary Interventions for Multiple Sclerosis. Cochrane Database Syst. Rev. 2012, 12, CD004192. [Google Scholar] [CrossRef]
  92. Swank, R.L.; Dugan, B.B. Effect of Low Saturated Fat Diet in Early and Late Cases of Multiple Sclerosis. Lancet 1990, 336, 37–39. [Google Scholar] [CrossRef]
  93. Swank, R.L.; Goodwin, J. Review of MS Patient Survival on a Swank Low Saturated Fat Diet 1 1 (For an Additional Perspective, See Editorial Opinions). Nutrition 2003, 19, 161. [Google Scholar] [CrossRef]
  94. Appel, L.J.; Van Horn, L. Did the PREDIMED Trial Test a Mediterranean Diet? N. Engl. J. Med. 2013, 368, 1353–1354. [Google Scholar] [CrossRef]
  95. Öztürk, Y.E.; Helvaci, E.M.; Sökülmez Kaya, P.; Terzi, M. Is Mediterranean Diet Associated with Multiple Sclerosis Related Symptoms and Fatigue Severity? Nutr. Neurosci. 2023, 26, 228–234. [Google Scholar] [CrossRef]
  96. O’Connor, L.E.; Biberstine, S.L.; Paddon-Jones, D.; Schwichtenberg, A.J.; Campbell, W.W. Adopting a Mediterranean-Style Eating Pattern with Different Amounts of Lean Unprocessed Red Meat Does Not Influence Short-Term Subjective Personal Well-Being in Adults with Overweight or Obesity. J. Nutr. 2018, 148, 1917–1923. [Google Scholar] [CrossRef] [PubMed]
  97. Black, L.J.; Baker, K.; Ponsonby, A.-L.; Van Der Mei, I.; Lucas, R.M.; Pereira, G. A Higher Mediterranean Diet Score, Including Unprocessed Red Meat, Is Associated with Reduced Risk of Central Nervous System Demyelination in a Case-Control Study of Australian Adults. J. Nutr. 2019, 149, 1385–1392. [Google Scholar] [CrossRef]
  98. Sand, I.K.; Benn, E.K.; Fabian, M.; Fitzgerald, K.C.; Digga, E.; Deshpande, R.; Miller, A.; Gallo, S.; Arab, L. Randomized-Controlled Trial of a Modified Mediterranean Dietary Program for Multiple Sclerosis: A Pilot Study. Mult. Scler. Relat. Disord. 2019, 36, 101403. [Google Scholar] [CrossRef]
  99. Serra-Majem, L.; Ribas, L.; Ngo, J.; Ortega, R.M.; García, A.; Pérez-Rodrigo, C.; Aranceta, J. Food, Youth and the Mediterranean Diet in Spain. Development of KIDMED, Mediterranean Diet Quality Index in Children and Adolescents. Public Health Nutr. 2004, 7, 931–935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Noormohammadi, M.; Ghorbani, Z.; Naser Moghadasi, A.; Saeedirad, Z.; Shahemi, S.; Ghanaatgar, M.; Rezaeimanesh, N.; Hekmatdoost, A.; Ghaemi, A.; Razeghi Jahromi, S. MIND Diet Adherence Might Be Associated with a Reduced Odds of Multiple Sclerosis: Results from a Case–Control Study. Neurol. Ther. 2022, 11, 397–412. [Google Scholar] [CrossRef] [PubMed]
  101. Van den Brink, A.C.; Brouwer-Brolsma, E.M.; Berendsen, A.A.; van de Rest, O. The Mediterranean, Dietary Approaches to Stop Hypertension (DASH), and Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) Diets Are Associated with Less Cognitive Decline and a Lower Risk of Alzheimer’s Disease—A Review. Adv. Nutr. 2019, 10, 1040–1065. [Google Scholar] [CrossRef] [Green Version]
  102. Sand, I.K.; Fitzgerald, K.C.; Gu, Y.; Brandstadter, R.; Riley, C.S.; Buyukturkoglu, K.; Leavitt, V.M.; Krieger, S.; Miller, A.; Lublin, F. Dietary Factors and MRI Metrics in Early Multiple Sclerosis. Mult. Scler. Relat. Disord. 2021, 53, 103031. [Google Scholar] [CrossRef]
  103. Challa, H.J.; Bandlamudi, M.; Uppaluri, K.R. Paleolithic Diet. In StatPearls [Internet]; StatPearls Publishing: Orlando, FL, USA, 2021. [Google Scholar]
  104. Learmonth, Y.; Dlugonski, D.; Pilutti, L.A.; Sandroff, B.M.; Klaren, R.; Motl, R.W. Psychometric Properties of the Fatigue Severity Scale and the Modified Fatigue Impact Scale. J. Neurol. Sci. 2013, 331, 102–107. [Google Scholar] [CrossRef]
  105. Irish, A.K.; Erickson, C.M.; Wahls, T.L.; Snetselaar, L.G.; Darling, W.G. Randomized Control Trial Evaluation of a Modified Paleolithic Dietary Intervention in the Treatment of Relapsing-Remitting Multiple Sclerosis: A Pilot Study. Degener. Neurol. Neuromuscul. Dis. 2017, 7, 1–18. [Google Scholar] [CrossRef] [Green Version]
  106. Bisht, B.; Darling, W.G.; Shivapour, E.T.; Lutgendorf, S.K.; Snetselaar, L.G.; Chenard, C.A.; Wahls, T.L. Multimodal Intervention Improves Fatigue and Quality of Life in Subjects with Progressive Multiple Sclerosis: A Pilot Study. Degener. Neurol. Neuromuscul. Dis. 2015, 5, 19–35. [Google Scholar] [CrossRef] [Green Version]
  107. Wahls, T.L.; Chenard, C.A.; Snetselaar, L.G. Review of Two Popular Eating Plans within the Multiple Sclerosis Community: Low Saturated Fat and Modified Paleolithic. Nutrients 2019, 11, 352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Chenard, C.A.; Rubenstein, L.M.; Snetselaar, L.G.; Wahls, T.L. Nutrient Composition Comparison between a Modified Paleolithic Diet for Multiple Sclerosis and the Recommended Healthy US-Style Eating Pattern. Nutrients 2019, 11, 537. [Google Scholar] [CrossRef] [Green Version]
  109. Jelinek, G. Overcoming Multiple Sclerosis: An Evidence-Based Guide to Recovery; Allen & Unwin: Crows Nest, Australia, 2010; ISBN 10: 1742371795/13: 9781742371795. [Google Scholar]
  110. Brenton, J.N.; Banwell, B.; Bergqvist, A.C.; Lehner-Gulotta, D.; Gampper, L.; Leytham, E.; Coleman, R.; Goldman, M.D. Pilot Study of a Ketogenic Diet in Relapsing-Remitting MS. Neurol.-Neuroimmunol. Neuroinflammation 2019, 6, e565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Di Majo, D.; Cacciabaudo, F.; Accardi, G.; Gambino, G.; Giglia, G.; Ferraro, G.; Candore, G.; Sardo, P. Ketogenic and Modified Mediterranean Diet as a Tool to Counteract Neuroinflammation in Multiple Sclerosis: Nutritional Suggestions. Nutrients 2022, 14, 2384. [Google Scholar] [CrossRef] [PubMed]
  112. Fitzgerald, K. Diet and Disease Modification in Multiple Sclerosis: A Nutritional Epidemiology Perspective; BMJ Publishing Group Ltd.: London, UK, 2018; Volume 89, ISBN 0022-3050. [Google Scholar]
  113. Cignarella, F.; Cantoni, C.; Ghezzi, L.; Salter, A.; Dorsett, Y.; Chen, L.; Phillips, D.; Weinstock, G.M.; Fontana, L.; Cross, A.H. Intermittent Fasting Confers Protection in CNS Autoimmunity by Altering the Gut Microbiota. Cell Metab. 2018, 27, 1222–1235.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ghezzi, L. Energy Restriction in People with Multiple Sclerosis: Is Time More Important than Calories? EBioMedicine 2022, 82, 104183. [Google Scholar] [CrossRef] [PubMed]
  115. Fitzgerald, K.C.; Bhargava, P.; Smith, M.D.; Vizthum, D.; Henry-Barron, B.; Kornberg, M.D.; Cassard, S.D.; Kapogiannis, D.; Sullivan, P.; Baer, D.J. Intermittent Calorie Restriction Alters T Cell Subsets and Metabolic Markers in People with Multiple Sclerosis. EBioMedicine 2022, 82, 104124. [Google Scholar] [CrossRef]
  116. Choi, I.Y.; Piccio, L.; Childress, P.; Bollman, B.; Ghosh, A.; Brandhorst, S.; Suarez, J.; Michalsen, A.; Cross, A.H.; Morgan, T.E. A Diet Mimicking Fasting Promotes Regeneration and Reduces Autoimmunity and Multiple Sclerosis Symptoms. Cell Rep. 2016, 15, 2136–2146. [Google Scholar] [CrossRef] [Green Version]
  117. McDougall, J.; Thomas, L.E.; McDougall, C.; Moloney, G.; Saul, B.; Finnell, J.S.; Richardson, K.; Petersen, K.M. Effects of 7 Days on an Ad Libitum Low-Fat Vegan Diet: The McDougall Program Cohort. Nutr. J. 2014, 13, 99. [Google Scholar] [CrossRef] [Green Version]
  118. Dean, C.; Parks, S.; Titcomb, T.J.; Arthofer, A.; Meirick, P.; Grogan, N.; Ehlinger, M.A.; Bisht, B.; Fox, S.S.; Daack-Hirsch, S. Facilitators of and Barriers to Adherence to Dietary Interventions Perceived by Women with Multiple Sclerosis and Their Support Persons. Int. J. MS Care 2022, 24, 235–241. [Google Scholar] [CrossRef] [PubMed]
  119. Snetselaar, L.G.; Cheek, J.J.; Fox, S.S.; Healy, H.S.; Schweizer, M.L.; Bao, W.; Kamholz, J.; Titcomb, T.J. Titcomb Efficacy of Diet on Fatigue and Quality of Life in Multiple Sclerosis. Neurology 2023, 100, e357. [Google Scholar] [CrossRef] [PubMed]
  120. Spain, R.I.; Piccio, L.; Langer-Gould, A.M. Langer-Gould The Role of Diet in Multiple Sclerosis. Neurology 2023, 100, 167. [Google Scholar] [CrossRef]
  121. Galus, W.; Walawska-Hrycek, A.; Rzepka, M.; Krzystanek, E. Vitamin D Supplementation Practices among Multiple Sclerosis Patients and Professionals. J. Clin. Med. 2022, 11, 7278. [Google Scholar] [CrossRef] [PubMed]
  122. Cantarel, B.L.; Waubant, E.; Chehoud, C.; Kuczynski, J.; DeSantis, T.Z.; Warrington, J.; Venkatesan, A.; Fraser, C.M.; Mowry, E.M. Gut Microbiota in Multiple Sclerosis: Possible Influence of Immunomodulators. J. Investig. Med. 2015, 63, 729–734. [Google Scholar] [CrossRef] [Green Version]
  123. Rumah, K.R.; Vartanian, T.K.; Fischetti, V.A. Oral Multiple Sclerosis Drugs Inhibit the in Vitro Growth of Epsilon Toxin Producing Gut Bacterium, Clostridium Perfringens. Front. Cell Infect. Microbiol. 2017, 7, 11. [Google Scholar] [CrossRef] [Green Version]
  124. Sadeghi Bahmani, D.; Kesselring, J.; Papadimitriou, M.; Bansi, J.; Pühse, U.; Gerber, M.; Shaygannejad, V.; Holsboer-Trachsler, E.; Brand, S. In Patients with Multiple Sclerosis, Both Objective and Subjective Sleep, Depression, Fatigue, and Paresthesia Improved after 3 Weeks of Regular Exercise. Front. Psychiatry 2019, 10, 265. [Google Scholar] [CrossRef] [Green Version]
  125. Sadeghi Bahmani, D.; Gonzenbach, R.; Motl, R.W.; Bansi, J.; Rothen, O.; Niedermoser, D.; Gerber, M.; Brand, S. Better Objective Sleep Was Associated with Better Subjective Sleep and Physical Activity; Results from an Exploratory Study under Naturalistic Conditions among Persons with Multiple Sclerosis. Int. J. Environ. Res. Public. Health 2020, 17, 3522. [Google Scholar] [CrossRef]
  126. Clauss, M.; Gérard, P.; Mosca, A.; Leclerc, M. Interplay between Exercise and Gut Microbiome in the Context of Human Health and Performance. Front. Nutr. 2021, 8, 637010. [Google Scholar] [CrossRef]
  127. Simpson, S.; Furlong, M.; Giebel, C. Exploring the Enablers and Barriers to Social Prescribing for People Living with Long-Term Neurological Conditions: A Focus Group Investigation. BMC Health Serv. Res. 2021, 21, 1230. [Google Scholar] [CrossRef]
  128. Keykhaei, F.; Norouzy, S.; Froughipour, M.; Nematy, M.; Saeidi, M.; Jarahi, L.; Amiri, F.; Malek Ahmadi, M.; Norouzy, A. Adherence to Healthy Dietary Pattern Is Associated with Lower Risk of Multiple Sclerosis. J. Cent. Nerv. Syst. Dis. 2022, 14, 11795735221092516. [Google Scholar] [CrossRef] [PubMed]
  129. Teng, M.; Bensmail, D.; Hanachi, M.; Haddad, R.; Hugeron, C.; Lansaman, T.; Levy, J. Nutritional Status in Patients with Advanced-stage Multiple Sclerosis. Eur. J. Neurol. 2022, 29, 1730–1740. [Google Scholar] [CrossRef] [PubMed]
  130. Robles, L.R.; De La Maza, B.P.; García, J.T.; Vieitez, J.J.G.; Gómez, M.J.F.; Mellado, I.B.; Pomar, M.D.B. Nutritional Profile of Multiple Sclerosis. Nutr. Hosp. Organo Soc. Esp. Nutr. Parenter. Enter. 2019, 36, 340–349. [Google Scholar] [CrossRef] [Green Version]
  131. Silveira, S.L.; Jeng, B.; Cutter, G.; Motl, R.W. Diet Quality Assessment in Wheelchair Users with Multiple Sclerosis. Nutrients 2021, 13, 4352. [Google Scholar] [CrossRef]
  132. Luca, M.; Ortega-Castro, N.; Patti, F. Paediatric Multiple Sclerosis: A Scoping Review of Patients’ and Parents’ Perspectives. Children 2021, 9, 11. [Google Scholar] [CrossRef] [PubMed]
  133. Ghai, S.; Kasilingam, E.; Lanzillo, R.; Malenica, M.; van Pesch, V.; Burke, N.C.; Carotenuto, A.; Maguire, R. Needs and Experiences of Children and Adolescents with Pediatric Multiple Sclerosis and Their Caregivers: A Systematic Review. Children 2021, 8, 445. [Google Scholar] [CrossRef]
  134. Langer-Gould, A.; Brara, S.M.; Beaber, B.E.; Koebnick, C. Childhood Obesity and Risk of Pediatric Multiple Sclerosis and Clinically Isolated Syndrome. Neurology 2013, 80, 548–552. [Google Scholar] [CrossRef] [Green Version]
  135. Milles, P.; De Filippo, G.; Maurey, H.; Tully, T.; Deiva, K. Obesity in Pediatric-Onset Multiple Sclerosis: A French Cohort Study. Neurol.-Neuroimmunol. Neuroinflammation 2021, 8, e1044. [Google Scholar] [CrossRef]
  136. Pakpoor, J.; Pakpoor, J. Childhood Obesity and Risk of Pediatric Multiple Sclerosis and Clinically Isolated Syndrome. Neurology 2013, 81, 1366. [Google Scholar] [CrossRef] [Green Version]
  137. Shahi, S.K.; Ghimire, S.; Lehman, P.; Mangalam, A.K. Obesity Induced Gut Dysbiosis Contributes to Disease Severity in an Animal Model of Multiple Sclerosis. Front. Immunol. 2022, 13, 966417. [Google Scholar] [CrossRef]
  138. Russell, R.D.; Langer-Gould, A.; Gonzales, E.G.; Smith, J.B.; Brennan, V.; Pereira, G.; Lucas, R.M.; Begley, A.; Black, L.J. Obesity, Dieting, and Multiple Sclerosis. Mult. Scler. Relat. Disord. 2020, 39, 101889. [Google Scholar] [CrossRef]
  139. Bruce, J.M.; Cozart, J.S.; Shook, R.P.; Ruppen, S.; Siengsukon, C.; Simon, S.; Befort, C.; Lynch, S.; Mahmoud, R.; Drees, B. Modifying Diet and Exercise in MS (MoDEMS): Study Design and Protocol for a Telehealth Weight Loss Intervention for Adults with Obesity & Multiple Sclerosis. Contemp. Clin. Trials 2021, 107, 106495. [Google Scholar] [CrossRef]
  140. Cercato, C.; Fonseca, F.A. Cardiovascular Risk and Obesity. Diabetol. Metab. Syndr. 2019, 11, 74. [Google Scholar] [CrossRef] [PubMed]
  141. Marrie, R.A.; Rudick, R.; Horwitz, R.; Cutter, G.; Tyry, T.; Campagnolo, D.; Vollmer, T. Vascular Comorbidity Is Associated with More Rapid Disability Progression in Multiple Sclerosis. Neurology 2010, 74, 1041–1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Schepici, G.; Silvestro, S.; Bramanti, P.; Mazzon, E. The Gut Microbiota in Multiple Sclerosis: An Overview of Clinical Trials. Cell Transplant. 2019, 28, 1507–1527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Sarkar, A.; Lehto, S.M.; Harty, S.; Dinan, T.G.; Cryan, J.F.; Burnet, P.W. Psychobiotics and the Manipulation of Bacteria–Gut–Brain Signals. Trends Neurosci. 2016, 39, 763–781. [Google Scholar] [CrossRef] [Green Version]
  144. Oroojzadeh, P.; Bostanabad, S.Y.; Lotfi, H. Psychobiotics: The Influence of Gut Microbiota on the Gut-Brain Axis in Neurological Disorders. J. Mol. Neurosci. 2022, 72, 1952–1964. [Google Scholar] [CrossRef] [PubMed]
  145. Kumar, N.; Sahoo, N.K.; Mehan, S. The Importance of Gut-Brain Axis and Use of Probiotics as a Treatment Strategy for Multiple Sclerosis. Mult. Scler. Relat. Disord. 2023, 71, 104547. [Google Scholar] [CrossRef]
  146. Hashemi, B.; Abdollahi, M.; Abbaspour-Aghdam, S.; Hazrati, A.; Malekpour, K.; Kafil, H.S.; Yousefi, M.; Roshangar, L.; Ahmadi, M. The Effect of Probiotics on Immune Responses and Their Therapeutic Application: A New Treatment Option for Multiple Sclerosis. Biomed. Pharmacother. 2023, 159, 114195. [Google Scholar] [CrossRef]
  147. Dziedzic, A.; Saluk, J. Probiotics and Commensal Gut Microbiota as the Effective Alternative Therapy for Multiple Sclerosis Patients Treatment. Int. J. Mol. Sci. 2022, 23, 14478. [Google Scholar] [CrossRef]
  148. Stephens, S.; Shams, S.; Lee, J.; Grover, S.A.; Longoni, G.; Berenbaum, T.; Finlayson, M.; Motl, R.W.; Yeh, E.A. Benefits of Physical Activity for Depression and Fatigue in Multiple Sclerosis: A Longitudinal Analysis. J. Pediatr. 2019, 209, 226–232.e2. [Google Scholar] [CrossRef] [PubMed]
  149. Parks, N.E.; Jackson-Tarlton, C.S.; Vacchi, L.; Merdad, R.; Johnston, B.C. Dietary Interventions for Multiple Sclerosis-related Outcomes. Cochrane Database Syst. Rev. 2020, 2020, CD004192. [Google Scholar] [CrossRef]
  150. Amatya, B.; Khan, F. Do Dietary Interventions Improve Health Outcomes in People with Multiple Sclerosis? A Cochrane Review Summary with Commentary. NeuroRehabilitation 2022, 50, 161–163. [Google Scholar] [CrossRef]
  151. Tredinnick, A.R.; Probst, Y.C. Evaluating the Effects of Dietary Interventions on Disease Progression and Symptoms of Adults with Multiple Sclerosis: An Umbrella Review. Adv. Nutr. 2020, 11, 1603–1615. [Google Scholar] [CrossRef] [PubMed]
  152. Galus, W.; Chmiela, T.; Walawska-Hrycek, A.; Krzystanek, E. Radiological Benefits of Vitamin D Status and Supplementation in Patients with MS—A Two-Year Prospective Observational Cohort Study. Nutrients 2023, 15, 1465. [Google Scholar] [CrossRef] [PubMed]
  153. Pediatric MS. Available online: https://www.nationalmssociety.org/For-Professionals/Clinical-Care/Managing-MS/Pediatric-MS (accessed on 7 May 2023).
  154. Greene, N.; Araujo, L.; Campos, C.; Dalglish, H.; Gibbs, S.; Yermilov, I. The Economic and Humanistic Burden of Pediatric-Onset Multiple Sclerosis. J. Health Econ. Outcomes Res. 2022, 9, 103–114. [Google Scholar] [CrossRef]
  155. Deiva, K. Pediatric Onset Multiple Sclerosis. Rev. Neurol. 2020, 176, 30–36. [Google Scholar] [CrossRef]
  156. Jeong, A.; Oleske, D.M.; Holman, J. Epidemiology of Pediatric-Onset Multiple Sclerosis: A Systematic Review of the Literature. J. Child Neurol. 2019, 34, 705–712. [Google Scholar] [CrossRef]
  157. Fernandez-Carbonell, C.; Charvet, L.E.; Krupp, L.B. Enhancing Mood, Cognition, and Quality of Life in Pediatric Multiple Sclerosis. Pediatr. Drugs 2021, 23, 317–329. [Google Scholar] [CrossRef]
  158. Benson, L.A.; Healy, B.C.; Gorman, M.P.; Baruch, N.F.; Gholipour, T.; Musallam, A.; Chitnis, T. Elevated Relapse Rates in Pediatric Compared to Adult MS Persist for at Least 6 Years. Mult. Scler. Relat. Disord. 2014, 3, 186–193. [Google Scholar] [CrossRef]
  159. Chitnis, T.; Aaen, G.; Belman, A.; Benson, L.; Gorman, M.; Goyal, M.S.; Graves, J.S.; Harris, Y.; Krupp, L.; Lotze, T. Improved Relapse Recovery in Paediatric Compared to Adult Multiple Sclerosis. Brain 2020, 143, 2733–2741. [Google Scholar] [CrossRef] [PubMed]
  160. Portaccio, E.; Amato, M.P. Cognitive Impairment in Multiple Sclerosis: An Update on Assessment and Management. NeuroSci 2022, 3, 667–676. [Google Scholar] [CrossRef]
  161. Luchesa Smith, A.; Benetou, C.; Bullock, H.; Kuczynski, A.; Rudebeck, S.; Hanson, K.; Crichton, S.; Mankad, K.; Siddiqui, A.; Byrne, S. Progress in the Management of Paediatric-Onset Multiple Sclerosis. Children 2020, 7, 222. [Google Scholar] [CrossRef]
  162. Immovilli, P.; De Mitri, P.; Bazzurri, V.; Vollaro, S.; Morelli, N.; Biasucci, G.; Magnifico, F.; Marchesi, E.; Lombardelli, M.L.; Gelati, L. The Impact of Highly Effective Treatment in Pediatric-Onset Multiple Sclerosis: A Case Series. Children 2022, 9, 1698. [Google Scholar] [CrossRef]
  163. Mrosková, S.; Klímová, E.; Majerníková, Ľ.; Tkáčová, Ľ. Quality of Life of Children and Adolescents with Multiple Sclerosis—A Literature Review of the Quantitative Evidence. Int. J. Environ. Res. Public. Health 2021, 18, 8645. [Google Scholar] [CrossRef]
  164. Brola, W.; Steinborn, B.; Żak, M.; Mazurkiewicz-Bełdzińska, M.; Jóźwiak, S.; Sobolewski, P.; Wilski, M.; Bilska, M.; Siedlarska, M.; Puzio-Bochen, I. The Clinical and Epidemiological Profile of Paediatric-Onset Multiple Sclerosis in Poland. J. Clin. Med. 2022, 11, 7494. [Google Scholar] [CrossRef]
  165. Lanzillo, R.; Chiodi, A.; Carotenuto, A.; Magri, V.; Napolitano, A.; Liuzzi, R.; Costabile, T.; Rainone, N.; Freda, M.F.; Valerio, P.; et al. Quality of Life and Cognitive Functions in Early Onset Multiple Sclerosis. Eur. J. Paediatr. Neurol. 2016, 20, 158–163. [Google Scholar] [CrossRef] [PubMed]
  166. Rainone, N.; Chiodi, A.; Lanzillo, R.; Magri, V.; Napolitano, A.; Morra, V.B.; Valerio, P.; Freda, M.F. Affective Disorders and Health-Related Quality of Life (HRQoL) in Adolescents and Young Adults with Multiple Sclerosis (MS): The Moderating Role of Resilience. Qual. Life Res. 2017, 26, 727–736. [Google Scholar] [CrossRef]
  167. Bigi, S.; Banwell, B. Pediatric Multiple Sclerosis. J. Child Neurol. 2012, 27, 1378–1383. [Google Scholar] [CrossRef]
  168. Brola, W.; Steinborn, B.; Niewada, M.; Mazurkiewicz-Bełdzińska, M.; Jóźwiak, S.; Sobolewski, P.; Żak, M.; Wilski, M.; Bilska, M.; Siedlarska, M. Pediatric-Onset Multiple Sclerosis in Poland: A Registry-Based Retrospective Cohort Study. Mult. Scler. Relat. Disord. 2022, 57, 103344. [Google Scholar] [CrossRef]
  169. Noncommunicable Diseases: Childhood Overweight and Obesity. Available online: https://www.who.int/news-room/questions-and-answers/item/noncommunicable-diseases-childhood-overweight-and-obesity (accessed on 7 May 2023).
  170. Obesity and Overweight. Available online: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (accessed on 7 May 2023).
  171. Mowry, E.M.; Krupp, L.B.; Milazzo, M.; Chabas, D.; Strober, J.B.; Belman, A.L.; McDonald, J.C.; Oksenberg, J.R.; Bacchetti, P.; Waubant, E. Vitamin D Status Is Associated with Relapse Rate in Pediatric-onset Multiple Sclerosis. Ann. Neurol. 2010, 67, 618–624. [Google Scholar] [CrossRef]
  172. Mosca, C.; Colucci, A.; Savoia, F.; Calì, C.; Del Bene, M.; Ranucci, G.; Maglione, A.; Pepe, A.; Morelli, A.; Vajro, P.; et al. Vitamin D Levels in the Pre- and Post-COVID-19 Pandemic Periods and Related Confinement at Pediatric Age. Nutrients 2023, 15, 2089. [Google Scholar] [CrossRef]
  173. Azary, S.; Schreiner, T.; Graves, J.; Waldman, A.; Belman, A.; Guttman, B.W.; Aaen, G.; Tillema, J.-M.; Mar, S.; Hart, J. Contribution of Dietary Intake to Relapse Rate in Early Paediatric Multiple Sclerosis. J. Neurol. Neurosurg. Psychiatry 2018, 89, 28–33. [Google Scholar] [CrossRef]
  174. Black, L.J.; Hetherton, S.; Forkan, M.; Gonzales, E.G.; Smith, J.B.; Daly, A.; Lucas, R.M.; Langer-Gould, A. An Exploratory Study of Diet in Childhood and Young Adulthood and Adult-Onset Multiple Sclerosis. Mult. Scler. J. 2021, 27, 1611–1614. [Google Scholar] [CrossRef]
  175. Duarte-Silva, E.; Meuth, S.G.; Peixoto, C.A. The Role of Iron Metabolism in the Pathogenesis and Treatment of Multiple Sclerosis. Front. Immunol. 2023, 14, 1019. [Google Scholar] [CrossRef] [PubMed]
  176. Weigel, K.J.; Lynch, S.G.; LeVine, S.M. Iron Chelation and Multiple Sclerosis. ASN Neuro 2014, 6, AN20130037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Pakpoor, J.; Seminatore, B.; Graves, J.S.; Schreiner, T.; Waldman, A.T.; Lotze, T.E.; Belman, A.; Greenberg, B.M.; Weinstock-Guttman, B.; Aaen, G. Dietary Factors and Pediatric Multiple Sclerosis: A Case-Control Study. Mult. Scler. J. 2018, 24, 1067–1076. [Google Scholar] [CrossRef]
  178. Van Rensburg, S.J.; Peeters, A.V.; Van Toorn, R.; Schoeman, J.; Moremi, K.E.; Van Heerden, C.J.; Kotze, M.J. Identification of an Iron-Responsive Subtype in Two Children Diagnosed with Relapsing-Remitting Multiple Sclerosis Using Whole Exome Sequencing. Mol. Genet. Metab. Rep. 2019, 19, 100465. [Google Scholar] [CrossRef]
  179. Mirza, A.I.; Zhu, F.; Knox, N.; Forbes, J.D.; Van Domselaar, G.; Bernstein, C.N.; Graham, M.; Marrie, R.A.; Hart, J.; Yeh, E.A. Metagenomic Analysis of the Pediatric-Onset Multiple Sclerosis Gut Microbiome. Neurology 2022, 98, e1050–e1063. [Google Scholar] [CrossRef]
  180. Tremlett, H.; Marrie, R.A. The Multiple Sclerosis Prodrome: Emerging Evidence, Challenges, and Opportunities. Mult. Scler. J. 2021, 27, 6–12. [Google Scholar] [CrossRef]
  181. Tremlett, H.; Fadrosh, D.W.; Faruqi, A.A.; Zhu, F.; Hart, J.; Roalstad, S.; Graves, J.; Lynch, S.; Waubant, E.; The US Network of Pediatric MS Centers. Gut Microbiota in Early Pediatric Multiple Sclerosis: A Case−Control Study. Eur. J. Neurol. 2016, 23, 1308–1321. [Google Scholar] [CrossRef] [Green Version]
  182. Liang, G.; Zhu, F.; Mirza, A.I.; Bar-Or, A.; Bernstein, C.N.; Bonner, C.; Forbes, J.D.; Graham, M.; Hart, J.; Knox, N.C. Stability of the Gut Microbiota in Persons with Paediatric-Onset Multiple Sclerosis and Related Demyelinating Diseases. Mult. Scler. J. 2022, 28, 1819–1824. [Google Scholar] [CrossRef] [PubMed]
  183. Pétrin, J.; Fiander, M.D.; Doss, P.M.I.A.; Yeh, E.A. A Scoping Review of Modifiable Risk Factors in Pediatric Onset Multiple Sclerosis: Building for the Future. Children 2018, 5, 146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Woolbright, E.; Koshiya, H.; Brenton, J.N. Body Size Perceptions & Diet Modification in Youth with Multiple Sclerosis. Mult. Scler. Relat. Disord. 2022, 58, 103402. [Google Scholar] [CrossRef]
  185. Stephens, S.; Schneiderman, J.E.; Finlayson, M.; Berenbaum, T.; Motl, R.W.; Yeh, E.A. Feasibility of a Theory-Informed Mobile App for Changing Physical Activity in Youth with Multiple Sclerosis. Mult. Scler. Relat. Disord. 2022, 58, 103467. [Google Scholar] [CrossRef] [PubMed]
  186. Qu, X.; Walsh, E.I.; Cherbuin, N.; Black, L.J. Mapping the Literature on Diet and Multiple Sclerosis: A Data-Driven Approach. Nutrients 2022, 14, 4820. [Google Scholar] [CrossRef]
  187. Ragnedda, G.; Leoni, S.; Parpinel, M.; Casetta, I.; Riise, T.; Myhr, K.-M.; Wolfson, C.; Pugliatti, M. Reduced Duration of Breastfeeding Is Associated with a Higher Risk of Multiple Sclerosis in Both Italian and Norwegian Adult Males: The EnvIMS Study. J. Neurol. 2015, 262, 1271–1277. [Google Scholar] [CrossRef]
  188. Holz, A.; Riefflin, M.; Heesen, C.; Riemann-Lorenz, K.; Obi, N.; Becher, H. Breastfeeding and Risk of Multiple Sclerosis: A Systematic Review and Meta-Analysis of Observational Studies. Neuroepidemiology 2022, 56, 391–401. [Google Scholar] [CrossRef]
  189. Hardy, D.; Chitnis, T.; Waubant, E.; Banwell, B. Preventing Multiple Sclerosis: The Pediatric Perspective. Front. Neurol. 2022, 13, 802380. [Google Scholar] [CrossRef]
  190. Brenton, J.N.; Piccio, L. Strengthening the Link: Diet Quality and Disability in MS; SAGE Publications Sage UK: London, UK, 2023; Volume 29, pp. 157–159. ISBN 1352-4585. [Google Scholar]
  191. Kim, C.H. Complex Regulatory Effects of Gut Microbial Short-Chain Fatty Acids on Immune Tolerance and Autoimmunity. Cell Mol. Immunol. 2023, 20, 341–350. [Google Scholar] [CrossRef]
  192. McDonald, J.; Graves, J.; Waldman, A.; Lotze, T.; Schreiner, T.; Belman, A.; Greenberg, B.; Weinstock-Guttman, B.; Aaen, G.; Tillema, J.-M. A Case-Control Study of Dietary Salt Intake in Pediatric-Onset Multiple Sclerosis. Mult. Scler. Relat. Disord. 2016, 6, 87–92. [Google Scholar] [CrossRef] [Green Version]
  193. Nourbakhsh, B.; Graves, J.; Casper, T.C.; Lulu, S.; Waldman, A.; Belman, A.; Greenberg, B.; Weinstock-Guttman, B.; Aaen, G.; Tillema, J.-M. Dietary Salt Intake and Time to Relapse in Paediatric Multiple Sclerosis. J. Neurol. Neurosurg. Psychiatry 2016, 87, 1350–1353. [Google Scholar] [CrossRef] [Green Version]
  194. Jakimovski, D.; Guan, Y.; Ramanathan, M.; Weinstock-Guttman, B.; Zivadinov, R. Lifestyle-Based Modifiable Risk Factors in Multiple Sclerosis: Review of Experimental and Clinical Findings. Neurodegener. Dis. Manag. 2019, 9, 149–172. [Google Scholar] [CrossRef] [PubMed]
  195. Sikes, E.M.; Motl, R.W.; Ness, J.M. Pediatric Multiple Sclerosis: Current Perspectives on Health Behaviors. Pediatr. Health Med. Ther. 2018, 9, 17–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Simpson-Yap, S.; De Brouwer, E.; Kalincik, T.; Rijke, N.; Hillert, J.A.; Walton, C.; Edan, G.; Moreau, Y.; Spelman, T.; Geys, L. Associations of Disease-Modifying Therapies with COVID-19 Severity in Multiple Sclerosis. Neurology 2021, 97, e1870–e1885. [Google Scholar] [CrossRef]
  197. Kerry, R.G.; Das, G.; Golla, U.; del Pilar Rodriguez-Torres, M.; Shin, H.-S.; Patra, J.K. Engineered Probiotic and Prebiotic Nutraceutical Supplementations in Combating Non-Communicable Disorders: A Review. Curr. Pharm. Biotechnol. 2022, 23, 72–97. [Google Scholar] [CrossRef]
  198. Ma, J.; Lyu, Y.; Liu, X.; Jia, X.; Cui, F.; Wu, X.; Deng, S.; Yue, C. Engineered Probiotics. Microb. Cell Factories 2022, 21, 72. [Google Scholar] [CrossRef]
  199. Pesce, M.; Seguella, L.; Del Re, A.; Lu, J.; Palenca, I.; Corpetti, C.; Rurgo, S.; Sanseverino, W.; Sarnelli, G.; Esposito, G. Next-Generation Probiotics for Inflammatory Bowel Disease. Int. J. Mol. Sci. 2022, 23, 5466. [Google Scholar] [CrossRef] [PubMed]
  200. Kohl, H.M.; Castillo, A.R.; Ochoa-Repáraz, J. The Microbiome as a Therapeutic Target for Multiple Sclerosis: Can Genetically Engineered Probiotics Treat the Disease? Diseases 2020, 8, 33. [Google Scholar] [CrossRef]
  201. Borody, T.; Leis, S.; Campbell, J.; Torres, M.; Nowak, A. Fecal Microbiota Transplantation (FMT) in Multiple Sclerosis (MS): 942. Off. J. Am. Coll. Gastroenterol. ACG 2011, 106, S352. [Google Scholar] [CrossRef]
  202. Engen, P.A.; Zaferiou, A.; Rasmussen, H.; Naqib, A.; Green, S.J.; Fogg, L.F.; Forsyth, C.B.; Raeisi, S.; Hamaker, B.; Keshavarzian, A. Single-Arm, Non-Randomized, Time Series, Single-Subject Study of Fecal Microbiota Transplantation in Multiple Sclerosis. Front. Neurol. 2020, 11, 978. [Google Scholar] [CrossRef]
  203. Al, K.F.; Craven, L.J.; Gibbons, S.; Parvathy, S.N.; Wing, A.C.; Graf, C.; Parham, K.A.; Kerfoot, S.M.; Wilcox, H.; Burton, J.P. Fecal Microbiota Transplantation Is Safe and Tolerable in Patients with Multiple Sclerosis: A Pilot Randomized Controlled Trial. Mult. Scler. J.–Exp. Transl. Clin. 2022, 8, 20552173221086664. [Google Scholar] [CrossRef] [PubMed]
  204. Matheson, J.-A.T.; Holsinger, R.D. The Role of Fecal Microbiota Transplantation in the Treatment of Neurodegenerative Diseases: A Review. Int. J. Mol. Sci. 2023, 24, 1001. [Google Scholar] [CrossRef]
  205. Martinelli, V.; Albanese, M.; Altieri, M.; Annovazzi, P.; Arabi, S.; Bucello, S.; Caleri, F.; Cerqua, R.; Costanzi, C.; Cottone, S. Gut-Oriented Interventions in Patients with Multiple Sclerosis: Fact or Fiction? Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 935–946. [Google Scholar] [CrossRef] [PubMed]
  206. Russell, R.D.; Black, L.J.; Begley, A. Nutrition Education Programs for Adults with Neurological Diseases Are Lacking: A Scoping Review. Nutrients 2022, 14, 1577. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Multifactor etiology of multiple sclerosis (MS) [4].
Figure 1. Multifactor etiology of multiple sclerosis (MS) [4].
Children 10 01022 g001
Figure 2. The blood-brain barrier consists of biological barriers formed by different cells at three key interfaces. The 1st layer is made up of microvascular endothelial cells that line the cerebral capillaries and permeate the brain and spinal cord. The 2nd barrier is made up of the epithelial cells of the choroid plexus. This layer, being more permeable to proteins thanks to the presence of a fenestrated endothelium below a cuboidal epithelium, may regulate brain permeability under conditions of gut inflammation. The 3rd barrier is situated below the dura mater and contributes little to blood-brain exchange due to its avascular nature and relatively small surface area compared to other barriers. CSF: Cerebrospinal fluid.
Figure 2. The blood-brain barrier consists of biological barriers formed by different cells at three key interfaces. The 1st layer is made up of microvascular endothelial cells that line the cerebral capillaries and permeate the brain and spinal cord. The 2nd barrier is made up of the epithelial cells of the choroid plexus. This layer, being more permeable to proteins thanks to the presence of a fenestrated endothelium below a cuboidal epithelium, may regulate brain permeability under conditions of gut inflammation. The 3rd barrier is situated below the dura mater and contributes little to blood-brain exchange due to its avascular nature and relatively small surface area compared to other barriers. CSF: Cerebrospinal fluid.
Children 10 01022 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mandato, C.; Colucci, A.; Lanzillo, R.; Staiano, A.; Scarpato, E.; Schiavo, L.; Operto, F.F.; Serra, M.R.; Di Monaco, C.; Napoli, J.S.; et al. Multiple Sclerosis—Related Dietary and Nutritional Issues: An Updated Scoping Review with a Focus on Pediatrics. Children 2023, 10, 1022. https://doi.org/10.3390/children10061022

AMA Style

Mandato C, Colucci A, Lanzillo R, Staiano A, Scarpato E, Schiavo L, Operto FF, Serra MR, Di Monaco C, Napoli JS, et al. Multiple Sclerosis—Related Dietary and Nutritional Issues: An Updated Scoping Review with a Focus on Pediatrics. Children. 2023; 10(6):1022. https://doi.org/10.3390/children10061022

Chicago/Turabian Style

Mandato, Claudia, Angelo Colucci, Roberta Lanzillo, Annamaria Staiano, Elena Scarpato, Luigi Schiavo, Francesca Felicia Operto, Maria Rosaria Serra, Cristina Di Monaco, Julia Sara Napoli, and et al. 2023. "Multiple Sclerosis—Related Dietary and Nutritional Issues: An Updated Scoping Review with a Focus on Pediatrics" Children 10, no. 6: 1022. https://doi.org/10.3390/children10061022

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop